Phase III
NCT01610284
[ ]
HR+: hormone receptors positive; HER2-: human epidermal growth factor receptor 2 negative; MBC: metastatic breast cancer; BC: breast cancer; PFS: progression free survival; CBR: clinical benefit rate; ORR: objective response rate; pCR: pathologic complete response; HR: hazard ratio.
The mTOR/PI3K/Akt pathway inhibitors can be divided into different categories according to the target in the pathway. Specific inhibitors have been developed to target all or specific isoforms of PI3K, mTORC1 and Akt [ 251 ].
Pan-PI3K inhibitors target all PI3K isoforms resulting in significant off-target effects. The main pan-PI3K inhibitors are buparlisib and pictilisib [ 252 ]. Multiple clinical trials have tested the effects of pan-PI3K inhibitors in luminal BC.
The phase III randomized double-blinded BELLE-2 trial compared buparlisib combined with fulvestrant, to fulvestrant monotherapy in luminal A advanced or metastatic BC patients [ 230 ]. The results of this trial showed a modest improvement in PFS when buparlisib was added to fulvestrant. Another phase III clinical trial (BELLE-3) studied the effects of buparlisib plus fulvestrant in luminal A advanced or metastatic BC patients with no benefits from endocrine therapy [ 231 ]. Though PFS was significantly improved with buparlisib, there were severe adverse effects such as hyperglycemia, dyspnea, or pleural effusion. Lastly, the phase II/III BELLE-4 clinical trial evaluated buparlisib plus paclitaxel in HER2-negative locally advanced or metastatic BC patients [ 232 ]. The addition of buparlisib to paclitaxel did not improve PFS in these patients. Thus, further studies on buparlisib in HR+ BC were not conducted. The phase II randomized, double-blinded FERGI clinical trial analyzed the effects of pictilisib plus fulvestrant in luminal A BC patients resistant to AI [ 233 ]. The addition of pictilisib to fulvestrant did not improve PFS. Moreover, severe adverse effects occurred when the dose of pictilisib was increased. These results were confirmed for pictilisib plus paclitaxel, as the phase II PEGGY study showed no benefit from pictilisib in PI3K-mutated HER2-negative BC patients [ 234 ].
Hence, pan-PI3K inhibitors are not optimal to treat HR+ BC due to their toxicity and lack of efficacy.
To sort out issues related to off-target effects and toxicities with pan-PI3K inhibitors, isoform-specific PI3K inhibitors have been developed. These isoform-specific PI3K inhibitors can specifically target the PI3K p110α, p110β, p110δ, and p110γ isoforms [ 252 ]. Multiple clinical trials have tested the effects of isoform-specific PI3K inhibitors.
PI3K p110α is the most commonly mutated isoform in BC [ 253 ]. Alpelisib is the first FDA-approved PI3K p110α isoform inhibitor. A phase Ib clinical trial tested the effects of alpelisib and letrozole in patients with ER+ metastatic BC refractory to endocrine therapy [ 235 ]. The clinical benefit of the alpselisib and letrozole combination was higher for patients with PI3K-mutated BC, but clinical activity was still observed in patients with non-mutated tumors. The phase III randomized SOLAR-1 clinical trial compared the effects of alpelisib plus fulvestrant to fulvestrant alone in luminal A advanced BC patients who received no benefits from prior endocrine therapy [ 236 ]. The addition of alpelisib improved PFS for patients with PI3K-mutated BC.
Taselisib targets the PI3K p110α, p110γ and p110δ isoforms [ 254 ]. Taselisib was tested in the SANDPIPER study, a phase III randomized clinical trial, in combination with fulvestrant in patients with ER+ metastatic BC resistant to AIs [ 238 ]. Although the addition of taselisib slightly improved PFS, further clinical trials with taselisib were interrupted since high rates of severe adverse events were detected.
mTORC1 inhibitors, such as everolimus, block the mTORC1 dependent phosphorylation of s6k1 [ 255 ]. The BOLERO-2 phase III randomized clinical trial investigated the effects of exemestane with or without everolimus in AI-resistant ER+ metastatic BC patients [ 240 ]. The combination of everolimus and exemestane improved PFS. The TAMRAD phase II randomized open-label study compared the effects of tamoxifen with or without everolimus in AI-resistant luminal A BC patients [ 241 ]. This study showed an improvement in overall survival (OS) when everolimus was given in combination with tamoxifen. The findings of these two clinical trials led to FDA approval of everolimus. More recently, the PrE0102 phase II randomized clinical trial showed that the addition of everolimus to fulvestrant improved PFS of patients with AI-resistant ER+ BC compared to fulvestrant alone [ 242 ].
Akt inhibitors target all Akt isoforms as Akt 1, 2, and 3 isoforms share very similar structures [ 256 ]. Capivasertib is the principal Akt inhibitor under investigation in different clinical trials. The FAKTION phase II multi-centered randomized clinical trial compared the effects of capivasertib plus fulvestrant to fulvestrant plus placebo in AI-resistant luminal A advanced BC patients [ 243 ]. PFS was significantly improved with the combination of capivasertib and fulvestrant in comparison with the placebo arm.
The AKT1 E17K activating mutation is the most common in Akt and occurs in approximately 7% of ER+ metastatic BC. This mutation in the Akt lipid-binding pocket leads to constitutive Akt activation by modifying its localization to the membrane [ 257 ]. A phase I study analyzed the effects of capivasertib alone or in combination with fulvestrant in a cohort of patients with AKT1 E17K mutation ER+ metastatic BC [ 244 ]. Capivasertib, in combination with fulvestrant, demonstrated clinically meaningful activity and better tolerability compared to capivasertib alone.
There are currently three CDK4/6 inhibitors approved to treat HR+/HER2- metastatic BC: palbociclib, ribociclib, and abemaciclib. They can be administered as first-line treatment combined with AIs or as second-line treatment combined with fulvestrant [ 258 ].
Palbociclib, a highly selective CDK4/6 inhibitor, is the first FDA-approved CDK4/6 inhibitor as first-line treatment combined with AIs for metastatic or advanced HR+ BC patients [ 259 ].
PALOMA-1 is an open-label, randomized phase II study that evaluated the effects of palbociclib in combination with letrozole vs. letrozole alone as first-line treatment for HR+ advanced BC patients [ 126 ]. The addition of palbociclib to letrozole significantly improved PFS in HR+ BC patients. A phase III study was performed (PALOMA-2) to confirm these findings and expand the efficacy and safety of palbociclib, [ 245 ]. This double-blinded clinical trial tested the combination of palbociclib and letrozole in postmenopausal BC patients without prior systemic therapy for advanced BC. The addition of palbociclib to letrozole significantly improved PFS and ORR.
Ribociclib is the second FDA-approved CDK4/6 inhibitor for first-line treatment in postmenopausal advanced BC patients in combination with AIs [ 260 ]. The phase III MONALEESA-2 clinical trial results showed improved PFS and ORR with the combination of ribociclib and letrozole in HR+ metastatic BC patients. The clinical benefits and manageable tolerability observed with ribociclib and letrozole are maintained with longer follow-up compared to letrozole alone [ 247 ].
Abemaciclib has been tested in the phase III randomized double-blinded MONARCH-3 study [ 250 ]. HR+ advanced BC patients with no prior systemic therapy received abemaciclib plus anastrozole or letrozole or AIs plus placebo in the control arm. PFS and ORR were significantly improved with the combination of abemaciclib and AIs.
As second-line treatment, palbociclib can be given in combination with fulvestrant in advanced or metastatic BC patients with disease progression after endocrine therapy [ 261 ]. This was confirmed in the phase III multi-centered randomized double-blinded PALOMA-3 trial [ 246 ]. BC patients who received palbociclib plus fulvestrant had significantly longer PFS compared to fulvestrant plus placebo.
The phase III MONALEESA-3 study tested the effects of ribociclib plus fulvestrant in patients with HR+ advanced BC who received prior endocrine therapy in the advanced setting [ 248 ]. The PFS and ORR were significantly improved when ribociclib was added to fulvestrant. Thus, ribociclib plus fulvestrant can be considered as second-line treatment for these BC patients.
Abemaciclib has been recently approved in combination with fulvestrant for HR+ advanced or metastatic BC patients with disease progression after endocrine therapy. This was based on the results of the phase III, double-blinded MONARCH 2 study [ 249 ]. The combination of abemaciclib and fulvestrant demonstrated a significant improvement of PFS and ORR compared to fulvestrant plus placebo in HR+ metastatic BC patients who experienced relapse or progression after prior endocrine therapy.
mTOR/PI3K/Akt inhibitors and CDK4/6 inhibitors show great promise for advanced HR+ BC resistant to endocrine therapy. To leverage the potential of these two types of therapies, some preclinical studies have evaluated a triple therapy combination including PI3K inhibitors, CDK4/6 inhibitors, and endocrine therapy (see the summarized table at the end of the manuscript) [ 262 ].
As mentioned in Section 3.2 , HER2+ BC is currently treated with specific HER2 targeting antibodies or tyrosine kinase inhibitors (TKIs), and more recently, with TDM-1, an antibody-drug conjugate. These treatments have greatly improved HER2+ BC survival. However, 25% of HER2+ BC patients will still develop resistance to anti-HER2 treatment. Hence, new therapeutic strategies are emerging, such as new antibodies targeting HER2, new TKIs, vaccines, and PI3K/mTOR and CDK4/6 inhibitors [ 263 ]. The most recent completed clinical trials on new strategies for HER2+ BC treatment are gathered in Table 2 .
Most recent completed clinical trials on emerging therapies for HER2+ breast cancer.
Targeted Therapy | Drug Name | Trial Number | Patient Population | Trial Arms | Outcomes |
---|---|---|---|---|---|
Antibodies drug conjugate (ADC) | Trastuzumab-deruxtcan (DS-8201a) | DESTINY-Breast01 Phase II NCT03248492 [ ] | HER2+ MBC Prior trastuzumab-emtansine treatment | Trastuzumab-deruxtcan monotherapy | PFS 16.4 months |
Trastuzumab-duocarmycin (SYD985) | Phase I dose-escalation and dose-expansion NCT02277717 [ ] | HER2+ Locally advanced or metastatic solid tumors | Trastuzumab-duocarmycin monotherapy | ORR 33% | |
Modified antibodies | Margetuxumab (MGAH22) | SOPHIA Phase III NCT02492711 [ ] | HER2+ Advanced or MBC Prior anti-HER2 therapies | Margetuximab + chemotherapy vs. trastuzumab + chemotherapy | PFS 5.8 months vs. 4.9 months (HR 0.76; = 0.03) OS 21.6 months vs. 19.8 months (HR 0.89; = 0.33) ORR 25% vs. 14% ( < 0.001) |
Tyrosine kinase inhibitors | Tucatinib | HER2CLIMB Phase II NCT02614794 [ ] | HER2+ Locally advanced or MBC Prior anti-HER2 therapies | Tucatinib + trastuzumab and capecitabine vs. placebo + trastuzumab and capecitabine | PFS 33.1% (7.8 months) vs. 12.3% (5.6 months) (HR 0.54; < 0.001) PFS 24.9% vs. 0% (HR 0.48; < 0.001) in brain metastases patients OS 44.9% vs. 26.6% (HR 0.66; = 0.005) |
Poziotinib | NOV120101-203 Phase II NCT02418689 [ ] | HER2+ MBC Prior chemotherapy and trastuzumab | Poziotinib monotherapy | PFS 4.04 months | |
HER2-derived peptide vaccine | E75 (NeuVax) | Phase I/II NCT00841399 NCT00854789 [ ] | HER2+ Node-positive or high-risk node-negative BC HLA2/3+ | E75 vaccination vs. non-vaccination | DFS 89.7% vs. 80.2% ( = 0.008) DFS 94.6% in optimal dosed patients ( = 0.005 vs. non-vaccination) |
GP2 | Phase II NCT00524277 [ ] | HER2 (IHC 1-3+) Disease free Node-positive or high-risk node-negative BC HLA2+ | GP2 + GM-CSF vs. GM-CSF alone | DFS 94% vs. 85% ( = 0.17) DFS 100% vs. 89% in HER2-IHC3+ ( = 0.08) | |
AE37 | Phase II NCT00524277 [ ] | HER2 (IHC 1-3+) Node-positive or high-risk node-negative BC | AE37 + GM-CSF vs. GM-CSF alone | DFS 80.8% vs. 79.5% ( = 0.70) DFS 77.2% vs. 65.7% ( = 0.21) HER2-low DFS 77.7% vs. 49.0% ( = 0.12) TNBC | |
PI3K inhibitors | Alpelisib | Phase I NCT02167854 [ ] | HER2+ MBC with a mutation Prior ado-trastuzumab emtansine and pertuzumab | Alpelisib + Trastuzumab + LJM716 | Toxicities limited drug delivery 72% for alpelisib 83% for LJM716 |
Phase I NCT02038010 [ ] | HER2+ MBC Prior trastuzumab-based therapy | Alpelisib + T-DM1 | PFS 8.1 months ORR 43% CBR 71% and 60% in prior T-DM1 patients | ||
Copanlisib | PantHER Phase Ib NCT02705859 [ ] | HER2+ Advanced BC Prior anti-HER2 therapies | Copanlisib + trastuzumab | Stable disease 50% | |
mTOR inhibitors | Everolimus | BOLERO-1 Phase III NCT00876395 [ ] | HER2+ Locally advanced BC No prior treatment | Everolimus + trastuzumab vs. placebo + trastuzumab | PFS 14.95 months vs. 14.49 months (HR 0.89; = 0.1166) PFS 20.27 months vs. 13.03 months (HR 0.66; = 0.0049) |
BOLERO-3 Phase III NCT01007942 [ ] | HER2+ Advanced BC Trastuzumab-resistant Prior taxane therapy | Everolimus + trastuzumab and vinorelbine vs. placebo + trastuzumab and vinorelbine | PFS 7.00 months vs. 5.78 months (HR 0.78; = 0.0067) | ||
CDK4/6 inhibitors | Palbociclib | SOLTI-1303 PATRICIA Phase II NCT02448420 [ ] | HER2+ ER+ or ER- MBC Prior standard therapy including trastuzumab | Palbociclib + trastuzumab | PFS 10.6 months (luminal) vs. 4.2 months (non-luminal) (HR 0.40; = 0.003) |
Ribociclib | Phase Ib/II NCT02657343 [ ] | HER2+ Advanced BC Prior treatment with trastuzumab, pertuzumab, and trastuzumab emtansine | Ribociclib + trastuzumab | PFS 1.33 months No dose-limiting toxicities | |
Abemaciclib | MonarcHER Phase II NCT02675231 [ ] | HER2+ Locally advanced or MBC Prior anti-HER2 therapies | Abemaciclib + trastuzumab and fulvestrant (A) vs. abemaciclib + trastuzumab (B) vs. standard-of-care chemotherapy + trastuzumab (C) | PFS 8.3 months (A) vs. 5.7 months (C) (HR 0.67; = 0.051) PFS 5.7 months (B) vs. 5.7 months (C) (HR 0.97; = 0.77) |
HER2+: human epidermal growth factor receptor 2 positive; ER+: estrogen receptor positive; HLA2/3: human leucocyte antigen 2/3; MBC: metastatic breast cancer; BC: breast cancer; PFS: progression free survival; CBR: clinical benefit rate; ORR: objective response rate; DFS: disease-free survival OS: overall survival GM-CSF: granulocyte macrophage colony-stimulated factor; HR: hazard ratio.
Novel types of antibodies have been developed to target HER2+ BC more efficiently. They can be divided into three categories: antibody-drug conjugates (ADC), modified antibodies, and bispecific antibodies.
ADCs are the combination of a specific monoclonal antibody and a cytotoxic drug that is released in the antigen-expressing cells [ 280 ]. The most common ADC is TDM-1, and the promising results with TDM-1 have led to the development of new ADCs.
Trastuzumab-deruxtecan (DS-8201a) is a HER2-targeting antibody (trastuzumab) linked to a DNA topoisomerase I inhibitor (deruxtecan) [ 281 ]. A phase I study demonstrated that DS-8201a had antitumor activity even with HER2 low-expressing tumors [ 282 ]. These results led to phase II and phase III clinical trials. The DESTINY-Breast01 clinical trial is an open-labeled, single-group, multicentered phase II study [ 264 ] was evaluated in HER2+ metastatic BC patients who received prior TDM-1 treatment. DS-8201a showed durable antitumor activity for these patients. Two phase III clinical trials are currently evaluating DS-8201a. DESTINY-Breast02 (ClinicalTrials.gov identifier: NCT03523585) is comparing DS-8201a to standard treatment (lapatinib or trastuzumab) in HER2+ metastatic BC patients previously treated with TDM-1. The DESTINY-Breast03 (ClinicalTrials.gov identifier: NCT03529110) trial is evaluating the effects of DS-8201a vs. TDM-1 in HER2+ metastatic BC patients with prior trastuzumab and taxane treatment.
Trastuzumab-duocarmycin (SYD985) is a HER-2 targeting antibody (trastuzumab) conjugate with a cleavable linker-duocarmycin payload that causes irreversible alkylation of the DNA in tumor cells leading to cell death [ 283 ]. A dose-escalation phase I study evaluated the effects of SYD85 in BC patients with variable HER2 status and refractory to standard cancer treatment [ 284 ]. Trastuzumab-duocarmycin showed clinical activity in heavily pretreated HER2+ metastatic BC patients, including TDM-1 resistant and HER2-low BC patients. After these promising results, a phase I expansion cohort study was performed on the same type of patients (heavily pretreated HER2+ or HER2-low BC patients) [ 265 ]. This study confirmed previous results on the efficacy of STD985. A phase III clinical trial (TULIP-ClinicalTrials.gov identifier: NCT03262935) is ongoing to compare SYD985 to the treatment chosen by the physician in HER2+ metastatic BC patients. Other ADCs are under clinical trials to test their safety and activity for HER2+ advanced BC patients. RC48 is an anti-HER2 antibody conjugated with monomethyl auristatin E that demonstrated promising efficacy and a manageable safety profile in an open-labeled, multicentered phase II study (ClinicalTrials.gov identifier: NCT02881138) [ 248 ]. PF06804103 conjugates an anti-HER2 monoclonal antibody and the cytotoxic agent, Aur0101. In a phase I study (ClinicalTrials.gov identifier: NCT03284723), PF06804103 showed manageable toxicity and promising antitumor activity [ 249 ]. XMT1522 showed encouraging results in a dose-escalation phase I study (ClinicalTrials.gov identifier: NCT02952729) [ 250 ]. MEDI4276, which targets two different HER2 epitopes and is linked to a microtubule inhibitor, showed promising clinical activity in a phase I study (ClinicalTrials.gov identifier: NCT02576548) [ 254 ] (see the summarized table at the end of the manuscript).
Margetuxumab (MGAH22) is a human/mouse chimeric IgG1 targeting HER2 monoclonal antibody. It is based on trastuzumab as it binds to the same epitope (subdomain IV or HER2 extracellular domain) but with an enhanced Fcγ domain. The substitution of five amino acids into the IgG1 Fc domain increases CD16A affinity, a receptor found on macrophages and natural-killer cells, and decreases CD32B affinity, leading to increased antibody-dependent cell-mediated cytotoxicity (ADCC) [ 285 ]. A phase I study evaluated margetuximab toxicity and tumor activity on HER2+ BC patients for whom no standard treatment was available [ 266 ]. This study showed promising single-agent activity of margetuximab as well as good tolerability. The phase III randomized open-labeled SOPHIA clinical trial (ClinicalTrials.gov Identifier: NCT02492711) compared margetuximab plus chemotherapy vs. trastuzumab plus chemotherapy in pretreated HER2+ advanced BC patients [ 286 ]. The combination of margetuximab and chemotherapy significantly improved the PFS of patients compared to trastuzumab plus chemotherapy. This study is still under investigation to collect data on OS (see the summarized table at the end of the manuscript).
Bispecific antibodies (BsAbs) can target two different epitopes in the same or different receptors by combining the functionality of two monoclonal antibodies [ 287 ]. MCLA-128 targets both HER2 and HER3 and have an enhanced ADCC activity [ 288 ]. A phase I/II study evaluated the safety, tolerability, and antitumor activity of MCLA-128 in patients with pretreated HER2+ metastatic BC.
Preliminary results showed encouraging clinical benefits of MCLA-128. An open-labeled, multicentered phase II study (ClinicalTrials.gov identifier: NCT03321981) is ongoing to evaluate the effects of MCLA-128 in combination with trastuzumab and chemotherapy in HER2+ metastatic BC patients.
ZW25 is a BsAb biparatopic that binds the IV and II subdomains of the HER2 extracellular domain, the binding epitopes of trastuzumab and pertuzumab, respectively [ 289 ]. The efficacy of ZW25 was evaluated in a phase I study given alone or in combination with chemotherapy in patients with advanced or metastatic HER2+ BC. The results of this study showed promising antitumor activity, and no-dose limiting was observed.
T-cell bispecific antibodies (TCBs) are another type of BsAbs recently developed. TCBs target the CD3-chain of the T-cell receptor and tumor-specific antigens, resulting in lymphocyte activation and tumor cell death [ 290 ].
GBR1302 targets both HER2 and CD3 receptors and directs T-cells to HER2+ tumor cells. A phase II study (ClinicalTrials.gov identifier: NCT03983395) is ongoing to determine the safety profile of the GBR1302 single agent in previously treated HER2+ metastatic BC. PRS-343 targets HER2 and the immune receptor CD137, a member of the tumor necrosis factor receptor family. Two clinical trials are investigating the effects of PRS-343 monotherapy (ClinicalTrials.gov identifier: NCT03330561) or in combination with other treatments (ClinicalTrials.gov identifier: NCT03650348) (see the summarized table at the end of the manuscript).
One of the strategies of immunotherapy is activating the patient’s immune system to kill cancer cells. Vaccination is an emerging approach to induce a tumor-specific immune response by targeting tumor-associated antigens, such as HER2 [ 291 ]. HER2-derived peptide vaccines comprise different parts of the HER2 molecule, such as E75 (extracellular domain), GP2 (transmembrane domain), and AE37 (intracellular domain) [ 292 ].
E75 (HER2/neu 369–377: KIFGSLAFL) has high affinity for HLA2 and HLA3 (human leucocyte antigen) that can stimulate T-cells against HER2 overexpressing tumor cells [ 293 ]. The efficacy of the E75 vaccine to prevent BC recurrence has been evaluated in a phase I/II study, in which high-risk HER2+ HLA2/3+ BC patients received the E75 vaccine [ 269 ]. The results demonstrated the safety and clinical efficacy of the vaccine as PFS was improved in the E75-vaccinated group compared to the unvaccinated group. Other clinical trials are currently investigating the efficacy of the E75 vaccine on HER2+ BC (see he summarized table at the end of the manuscript).
GP2 (654-662: IISAVVGIL) is a subdominant epitope with poor affinity for HLA2 [ 294 ]. A phase I trial evaluating the effects of a GP2 vaccine in disease-free BC patients showed that it was safe and tolerable with HER2-specific immune response [ 295 ]. The GP2 vaccine has been tested in a randomized, open-labeled phase II study to prevent BC recurrence. The patients that received the GP2 vaccine had HER2+ and HLA2+ BC and were disease-free with a high risk of recurrence at the time of the study [ 270 ]. The results demonstrated that the GP2 vaccine was safe and clinically beneficial for patients with HER2+ BC who received the full vaccine series.
AE37 (Ii-key hybrid of MHC II peptide AE36 (HER2/neu 776–790)) can stimulate CD8+ and CD4+ cells. A randomized, single-blinded phase II study evaluated the effects of an AE37 vaccine to prevent BC recurrence. Patients with a high risk of recurrence and HER2+ BC received the AE37 vaccine [ 271 ]. The vaccination demonstrated no benefit in the overall intention-to-treat analysis, a method that considers the randomized treatment to avoid bias happening after the randomization [ 296 ]. However, the study showed that the AE37 vaccine was safe, and results suggested that it could be effective for HER2-low BC, such as TNBC.
As previously described in this review (see Section 3.2.2 Tyrosine kinase inhibitors (TKIs)), TKIs are small molecules targeting the HER2 intracellular catalytic domain [ 159 ]. New TKIs have been developed with better efficacy and less toxicity in the treatment of HER2+ metastatic BC, such as tucatinib and poziotinib.
Tucatinib is a TKI with high selectivity for HER2, leading to less EGFR-related toxicities, common with other HER TKIs [ 297 ]. A phase I dose-escalation trial evaluated the combination of tucatinib and trastuzumab in BC patients with progressive HER2+ brain metastases [ 298 ]. This study showed preliminary evidence of tucatinib efficacy and tolerability in these patients. Tucatinib was also tested in combination with TDM-1 in a phase Ib trial in HER2+ metastatic BC patients with heavy pre-treatment [ 299 ]. The combination of tucatinib and TDM-1 showed acceptable toxicity and antitumor activity in these patients. Tucatinib was FDA approved in combination with trastuzumab and capecitabine for patients with advanced or metastatic HER2+ BC who received prior anti-HER2 in the metastatic setting [ 300 ]. This was based on the results of the phase II HER2CLIMB clinical trial, where HER2+ metastatic BC patients received tucatinib or placebo in combination with trastuzumab and capecitabine [ 267 ]. The addition of tucatinib to trastuzumab and capecitabine improved PFS and OS of heavily pretreated HER2+ metastatic BC patients.
Poziotinib is a pan-HER kinase inhibitor that irreversibly inhibits the HER family members’ kinase activity [ 301 ]. A phase I study evaluated the efficacy and tolerability of poziotinib in advanced solid tumors. The results showed encouraging antitumor activity against different types of HER2+ cancers as poziotinib was safe and well-tolerated by the patients [ 302 ]. The phase II NOV120101-203 study evaluated the safety and efficacy of poziotinib monotherapy in heavily pretreated HER2+ metastatic BC patients [ 268 ]. Poziotinib showed meaningful activity in these patients with no severe toxicities.
As mentioned in the previous Section 4.1 , mTOR/PI3K inhibitors and CDK4/6 inhibitors have been evaluated as potential new strategic therapies for HR+ BC resistant to endocrine therapy. The mTOR/PI3K signaling pathway and CDK4/6 also play a role in the mechanisms leading to treatment resistance in HER2+ BC [ 303 ]. Thus, targeting them with mTOR/PI3K and CDK4/6 inhibitors is also being investigated in HER2+ BC subtype.
Alpelisib and taselisib are PI3K isoform-specific inhibitors that were also evaluated in HR+ BC [ 235 , 236 , 238 , 253 , 254 ]. A phase I study evaluated alpelisib in combination with trastuzumab and LJM716 (a HER3-targeted antibody) in patients with PI3KCA mutant HER2+ metastatic BC [ 272 ]. Unfortunately, the results of this study were limited by high gastrointestinal toxicity. Another phase I study tested alpelisib in combination with TDM-1 in HER2+ metastatic BC patients pretreated with trastuzumab [ 273 ]. The combination of alpelisib and TDM-1 demonstrated tolerability and antitumor activity in trastuzumab-resistant HER2+ metastatic BC patients. Taselisib is being tested in an ongoing phase Ib dose-escalation trial in combination with anti-HER2 therapies (trastuzumab, pertuzumab and TDM-1) in HER2+ advanced BC patients (ClinicalTrials.gov identifier: NCT02390427).
Copanlisib is a highly selective and potent pan-class I PI3K inhibitor [ 304 ]. A phase Ib (PantHER) study evaluated the tolerability and activity of copanlisib in combination with trastuzumab in heavily pretreated HER2+ metastatic BC patients [ 274 ]. The combination of copanlisib and trastuzumab was safe and tolerable. Preliminary evidence of tumor stability was observed in these patients.
Everolimus is a mTORC1 inhibitor also tested in HR+ BC [ 240 , 241 , 242 ]. Everolimus was tested in phase III clinical trials, in combination with trastuzumab and docetaxel (BOLERO-1), or in combination with trastuzumab and vinorelbine (BOLERO-3) in trastuzumab-resistant advanced HER2+ BC [ 275 , 276 ]. Unfortunately, results showed an increase of adverse effects with everolimus. Moreover, the BOLERO-1 clinical trial showed no improvement in PFS with the combination of trastuzumab and everolimus. By contrast, PFS was significantly longer when everolimus was added to vinorelbine in BOLERO-3. A study analyzing the molecular alterations found in patients in the BOLERO-1 and BOLERO-3 clinical trials demonstrated that HER2+ BC patients could derive more benefit from everolimus if the tumors had PI3KCA mutations, PTEN loss or a hyperactive PI3K pathway [ 305 ].
Palbociclib, ribociclib and abemaciclib are CDK4/6 inhibitors that have been FDA approved to treat HR+ BC as first-line treatments [ 247 , 250 , 259 ]. They have also been evaluated in multiple clinical trials for advanced HER2+ BC. Palbociclib has been tested in combination with trastuzumab in the phase II SOLTI-1303 PATRICIA clinical trial in heavily pretreated advanced HER2+ BC patients [ 277 ]. Palbociclib combined with trastuzumab demonstrated safety and encouraging survival outcomes in these patients. Palbociclib has also been evaluated in combination with TDM-1 in HER2+ advanced BC patients pretreated with trastuzumab and taxane therapy [ 306 ]. The results of this phase I/Ib study showed safety, tolerability, and antitumor activity in these patients.
Ribociclib was evaluated in a phase Ib/II trial in combination with trastuzumab to treat advanced HER2+ BC patients previously treated with multiple anti-HER2 therapies [ 278 ]. The combination of ribociclib and trastuzumab was safe, but there was limited activity in heavily pretreated patients. The conclusions of this study suggest that CDK4/6 inhibitor/anti-HER2 combination should be administered in patients with few previous therapies.
Abemaciclib has been tested in the phase II randomized open-labeled MonarcHER trial in combination with trastuzumab with or without fulvestrant vs. trastuzumab with standard chemotherapy in HR+/HER2+ BC patients [ 279 ]. The combination of abemaciclib, trastuzumab, and fulvestrant significantly improved PFS in these patients, with a tolerable safety profile.
There are multiple ongoing clinical trials for advanced HER2+ BC testing the combination of palbociclib, trastuzumab, pertuzumab, and anastrozole (ClinicalTrials.gov identifier: NCT03304080); or palbociclib and trastuzumab plus letrozole (ClinicalTrials.gov identifier: NCT03054363). Preliminary results are expected around July 2021 and March 2022, respectively (see he summarized table at the end of the manuscript).
A great proportion of HER2+ BC patients develop resistance to traditional anti-HER2 therapies, and 40–50% of patients with advanced HER2+ BC develop brain metastases [ 307 ]. Thus, developing new therapies to overcome resistance is essential. The therapeutic strategies that have been described in this section provide new hope for HER2+ BC patients, especially for advanced or metastatic HER2+ BC patients.
TNBC is the most aggressive BC subtype. The fact that TNBC lacks ER and PR expression and does not overexpress HER2, combined with its high heterogeneity, has contributed to the difficulties in developing efficient therapies [ 308 ]. Thus, multiple strategic therapies have been developed to treat all TNBC subtypes. These include conjugated antibodies, targeted therapy, and immunotherapy. An overview of the most recent and completed clinical trials on emerging therapies for TNBC is presented in Table 3 .
Most recent completed clinical trials on emerging therapies for TNBC.
Targeted Therapy | Drug Name | Trial Number | Patient Population | Trial Arms | Outcomes |
---|---|---|---|---|---|
Antibodies Drug Conjugate | Sacituzumab govitecan | ASCENT Phase III NCT02574455 [ ] | TNBC MBC Prior standard treatment | Sacituzumab govitecan vs. single-agent chemotherapy | PFS 5.6 months vs. 1.7 months (HR 0.41; < 0.001) PFS 12.1 months vs. 6.7 months (HR 0.48; < 0.001) |
VEGF inhibitors | Bevacizumab | BEATRICE Phase III NCT00528567 [ ] | Early TNBC Surgery | Bevacizumab + chemotherapy vs. chemotherapy alone | IDFS 80% vs. 77% OS 88% vs. 88% |
CALGB 40603 Phase II NCT00861705 [ ] | TNBC Stage II to III | Bevacizumab + chemotherapy vs. chemotherapy alone or Carboplatin + chemotherapy vs. chemotherapy alone | pCR 59% vs. 48% ( = 0.0089) (Bevacizumab) pCR 60% vs. 44% ( = 0.0018) (Carboplatin) | ||
EGFR inhibitors | Cetuximab | TBCRC 001 Phase II NCT00232505 [ ] | TNBC MBC | Cetuximab + carboplatin | Response < 20% TTP 2.1 months |
Phase II NCT00463788 [ ] | TNBC MBC Prior chemotherapy treatment | Cetuximab + cisplatin vs. cisplatin alone | ORR 20% vs. 10% ( = 0.11) PFS 3.7 months vs. 1.7 months (HR 0.67; = 0.032) OS 12.9 months vs. 9.4 months (HR 0.82; = 0.31) | ||
mTORC1 inhibitors | Everolimus | Phase II NCT00930930 [ ] | TNBC Stage II or III Neoadjuvant treatment | Everolimus + cisplatin and paclitaxel vs. placebo + cisplatin and paclitaxel | pCR 36% vs. 49% |
Akt inhibitors | Ipatasertib | LOTUS Phase II NCT02162719 [ ] | TNBC Locally advanced or MBC No prior sytemic therapy | Ipatasertib + paclitaxel vs. placebo + paclitaxel | PFS 6.2 months vs. 4.9 months (HR 0.60; = 0.037) PFS 6.2 months vs. 3.7 moths (HR 0.58; = 0.18) in PTEN-low patients |
FAIRLANE Phase II NCT02301988 [ ] | Early TNBC Neoadjuvant treatment | Ipatasertib + paclitaxel vs. placebo + paclitaxel | pCR 17% vs. 13% pCR 16% vs. 13% PTEN-low patients pCR 18% vs. 12% PIK3CA/AKT1/PTEN-altered patients | ||
Capivasertib | PAKT Phase II NCT02423603 [ ] | TNBC MBC No prior chemotherapy treatment | Capivasertib + paclitaxel vs. placebo + paclitaxel | PFS 5.9 months vs. 12.6 months (HR 0.61; = 0.04) | |
Androgen receptor inhibitors | Bicalutamide | Phase II NCT00468715 [ ] | HR- AR+ or AR- MBC | Bicalutamide monotherapy | CBR 19% PFS 12 weeks |
Enzalutamide | Phase II NCT01889238 [ ] | TNBC AR+ Locally advanced or MBC | Enzalutamide monotherapy | CBR 25% OS 12.7 months | |
CYP17 inhibitors | Abiraterone acetate | UCBG 12-1 Phase II NCT01842321 [ ] | TNBC AR+ Locally advanced or MBC Centrally reviewed Prior chemotherapy | Abiraterone acetate + prednisone | CBR 20% ORR 6.7% PFS 2.8 months |
Anti-PDL1 antibodies | Atezolizumab | Impassion 130 Phase III NCT02425891 [ ] | TNBC Locally advanced or MBC No prior treatment | Atezolizumab + nab-paclitaxel vs. placebo + nab-paclitaxel | OS 21.0 months vs. 18.7 months (HR 0.86; = 0.078) OS 25.0 months vs. 18.0 months (HR 0.71, 95% CI 0.54–0.94)) in PDL-1+ patients |
Impassion 031 Phase III NCT03197935 [ ] | TNBC Stage II to III No prior treatment | Atezolizumab + chemotherapy vs. placebo + chemotherapy | pCR 95% vs. 69% = 0.0044 | ||
Durvalumab | GeparNuevo Phase II NCT02685059 [ ] | TNBC MBC Stromal tumor-infiltrating lymphocyte (sTILs) | Durvalumab vs. placebo | pCR 53.4% vs. 44.2% pCR 61.0% vs. 41.4% in window cohort | |
SAFIRO BREAST-IMMUNO Phase II NCT02299999 [ ] | HER2- MBC Prior chemotherapy | Durvalumab vs. maintenance chemotherapy | HR of death 0.37 for PDL-1+ patients HR of death 0.49 for PDL-1- patients | ||
Phase I NCT02484404 [ ] | Recurrent women’s cancers including TNBC | Durvalumab + cediranib + olaparib | Partial response 44% CBR 67% | ||
Avelumab | JAVELIN Phase Ib NCT01772004 [ ] | MBC Prior standard-of-care therapy | Avelumab monotherapy | ORR 3.0% overall ORR 5.2% in TNBC ORR 16.7% in PDL-1+ vs. 1.6% in PDL-1- overall ORR 22.2.% in PDL-1+ vs. 2.6% in PDL-1- in TNBC | |
Anti-PD1 antibodies | Pembrolizumab | KEYNOTE-086 Phase II NCT02447003 [ ] | TNBC MBC Prior or no prior systemic therapy | Pembrolizumab monotherapy | Previously treated patients: ORR 5.3% overall ORR 5.7% PDL-1+ patients PFS 2.0 months OS 9.0 months Non-previously pretreated: ORR 21.4% PFS 2.1 months OS 18.0 months |
KEYNOTE-119 Phase III NCT02555657 [ ] | TNBC MBC Prior systemic therapy | Pembrolizumab vs. chemotherapy | OS 12.7 months vs. 11.6 months (HR 0.78; = 0.057) in PDL1+ patients OS 9.9 months vs. 10.8 months (HR 0.97, 95% CI 0.81–1.15) | ||
KEYNOTE-355 Phase III NCT02819518 [ ] | TNBC MBC No prior systemic therapy | Pembrolizumab + chemotherapy vs. placebo + chemotherapy | PFS 9.7 months vs. 5.6 months (HR 0.65; = 0.0012) in PDL-1+ patients PFS 7.6 months vs. 5.6 months (HR 0.74; = 0.0014) | ||
KEYNOTE-522 Phase III NCT03036488 [ ] | Early TNBC Stage II to III No prior treatment | Pembrolizumab + paclitaxel and carboplatin vs. placebo + paclitaxel and carboplatin | pCR 64.8% vs. 51.2 % ( < 0.001) | ||
Anti-CDL4 antibodies | Tremelimumab | Phase I [ ] | Incurable MBC | Tremelimumab + radiotherapy | OS 50.8 months |
Vaccines | PPV | Phase II UMIN000001844 [ ] | TNBC MBC Prior systemic therapy | PPV vaccine | PFS 7.5 months OS 11.1 months |
STn-KLH | Phase III NCT00003638 [ ] | MBC Prior chemotherapy Partial or complete response | STn-KLH vaccine vs. non-vaccine | TTP 3.4 months vs. 3.0 months |
TNBC: triple negative breast cancer; HER2: human epidermal growth factor receptor; HR: hormonal receptor; MBC: metastatic breast cancer; BC: breast cancer; AR: androgen receptor; PPV: personalized peptide vaccine; PFS: progression free survival; CBR: clinical benefit rate; ORR: objective response rate; IDFS: invasive disease-free survival; OS: overall survival; TTP: time to progression; pCR: pathologic complete response; HR: hazard ratio.
Antibody drug conjugates (ADCs) deliver a cytotoxic drug into the tumor cell by the specific binding of an antibody to a surface molecule [ 280 ]. Multiple ADCs have been investigated in TNBC such as sacituzumab govitecan, ladiratuzumab vedotin, or trastuzumab deruxtecan.
Sacituzumab govitecan combines an antibody targeting trophoblast antigen 2 (Trop-2) and a topoisomerase I inhibitor SN-38 [ 334 ]. Trop-2, a CA 2+ signal transducer, is expressed in 90% of TNBCs and is associated with poor prognosis [ 335 , 336 ]. A single-arm, multicentered phase I/II study evaluated sacituzumab govitecan in heavily pretreated metastatic TNBC patients [ 336 , 337 ]. The efficacy and safety of scituzumab govitecan was shown in these patients, as it was associated with durable objective response. Based on these results, a randomized phase III trial (ASCENT) tested sacituzumab govitecan compared to single-agent chemotherapy chosen by the physician in patients with relapsed or refractory metastatic TNBC [ 309 ]. Sacituzumab govitecan significantly improved PFS and OS of metastatic TNBC patients compared to chemotherapy.
Ladiratuzumab vedotin is composed of a monoclonal antibody targeting the zinc transporter LIV-1 and a potent microtubule disrupting agent, monoethyl auristatin E (MMAE) [ 338 ]. LIV-1 is a transmembrane protein with potent zinc transporter and metalloproteinase activity, expressed in more than 70% of metastatic TNBC tumors [ 339 ]. All clinical trials investigating ladiratuzumab vedotin are still ongoing. A dose-escalation phase I study is evaluating the safety and efficacy of ladiratuzumab vedotin in heavily pretreated metastatic TNBC patients (ClinicalTrials.gov identifier: NCT01969643). Preliminary results showed encouraging antitumor activity and tolerability of ladiratuzumab vedotin with an objective response rate of 32% [ 340 ]. The estimated study completion date is June 2023. Two phase Ib/II trials are testing ladiratuzumab vedotin in combination with immunotherapy agents in metastatic TNBC patients, such as pembrolizumab (ClinicalTrials.gov Identifier: NCT03310957) with expected preliminary results in February 2022, or in combination with multiple immunotherapy-based treatments (ClinicalTrials.gov Identifier: NCT03424005) with expected preliminary results in January 2023.
Trastuzumab deruxtecan is an ADC developed as a treatment for metastatic HER2+ BC patients. Its mechanism of action is described in Section 3.2 . Even though trastuzumab deruxtecan was developed to treat HER2+ BC, it showed antitumor activity in HER2-low tumors in a phase I study [ 282 ]. Based on these results, an ongoing open-labeled, multicentered phase III study (ClinicalTrials.gov Identifier: NCT03734029) is recruiting patients with HER2-low metastatic BC to test trastuzumab deruxtecan vs. standard treatment chosen by the physician. Preliminary results are expected in January 2023 (see Table 4 ).
Ongoing clinical trials on emerging therapies for BC treatment for all BC molecular subtypes.
Targeted Therapy | Drug Name | Patient Population | Trial Arms | Outcome Measures | Status | Trial |
---|---|---|---|---|---|---|
PI3K inhibitors | Copanlisib | HR+/HER2- Postmenopausal Invasive BC Stage I to IV | Copanlisib + letrozole and palbocilib vs. copanlisib + letrozole vs. letrozole + palbociclib | pCR ORR DLT | Active, not recruiting | Phase I/II NCT03128619 |
HR+/HER2- MBC Stage IV | Copanlisib + fulvestrant vs. fulverstant alone | PFS ORR | Recruiting | Phase I/II NCT03803761 | ||
HER2+ PIK3CA or PTEN mutated MBC Stage IV | Copanlisib + trastuzumab + pertuzumab vs. trastuzumab + pertuzumab | PFS OS DLT | Recruiting | Phase Ib/II NCT04108858 | ||
TNBC MBC Unresectable BC Stage III to IV | Copanlisib + eribulin vs. eribulin alone | MTD PFS ORR CBR | Recruiting | Phase I/II NCT04345913 | ||
Taselisib | HER2+ MBC Recurrent BC | Taselisib + TDM-1 vs. taselisib + TDM-1 and pertuzumab vs. taselisib + pertuzumab and trastuzumab vs. taselisib + pertuzumab and trastuzumab and paclitaxel | MTD PFS CBR | Active, not recruiting | Phase Ib NCT02390427 | |
mTOR inhibitors | Everolimus | TNBC Advanced BC Prior systemic treatment | Everolimus + caroboplatin vs. carboplatin alone | PFS ORR OS CBR | Recruiting | Phase II NCT02531932 |
Akt inhibitors | Capivasertib | HR+/HER2- Locally advanced or MBC Prior systemic treatment | Capivasertib + palbociclib and fulvesrant vs. pplacebo + palbociclib and fulvesrant | DLT PFS ORR CBR OS | Recruiting | Phase Ib/III NCT04862663 |
HR+/HER2- Locally advanced or MBC Prior systemic treatment | Capivasertib + fulvesrant vs. pplacebo + fulvesrant | PFS ORR CBR OS | Recruiting | Phase III NCT04305496 | ||
TNBC Locally advanced or MBC No prior systemic treatment | Capivasertib + paclitaxel vs. placebo + paclitaxel | PFS ORR CBR OS | Recruiting | Phase III NCT03997123 | ||
Ipatasertib | ER+/HER2- Post-menopausal Prior CDK4/6 inhibitors and AIs | Ipatasertib + fulvestrant verus placebo + fulvestrant | PFS ORR CBR OS | Recruiting | Phase III NCT04650581 | |
HR+/HER2- Post-menopausal Locally advanced or MBC Prior systemic treatment | Ipatasertib + fulverstrant vs. ipatasertib + AI vs. ipatasertib + fulvestrant and palbociclib | PFS ORR OS | Recruiting | Phase III NCT03959891 | ||
HER2+ PIK3CA mutated Locally advanced or MBC Prior systemic treatment | Ipatasertib + trastuzumab and pertuzumab | Safety and tolerability PFS ORR CBR | Recruiting | Phase Ib NCT04253561 | ||
TNBC MBC Stage IV No prior treatment | Ipatasertib + carboplatin and paclitaxel vs. ipatasertib + carboplatin vs. ipatasertib + capecitabine and atezolizumab | PFS CBR OS TTF | Recruiting | Phase I/Ib NCT03853707 | ||
TNBC Locally advanced or MBC Prior systemic treatment | Ipatasertib + capecitabine vs. ipatasertib + eribulin vs. ipatasertib + carboplatin and gemcitabine | PFS ORR CBR OS TTR | Recruiting | Phase IIa NCT04464174 | ||
CDK4/6 inhibitors | Ribociclib | HR+/HER2- PIK3CA mutated Postmenopausal Locally advanced or MBC No prior systemic treatment | Ribociclib + letrozole | TTP CBR | Active, not recruiting | Phase III NCT03439046 |
HR+/HER2- MBC Prior systemic treatment | Ribociclib + (anti-hormonal treatment) anastrozole and exemestane and letrozole and fulvestrant vs. anti-hormonal treatment alone | PFS CBR OS | Recruiting | Phase II NCT03913234 | ||
HR+/HER2- Early BC No prior endocrine therapy | Ribociclib + endocrine therapy vs. endocrine therapy alone | IDFS RFS DDFS OS | Recruiting | Phase III NCT03701334 | ||
HR+/HER2- Locally advanced or MBC No prior systemic treatment | Ribociclib monotherapy | ORR PFS CBR TTP | Active, not recruiting | Phase II NCT03822468 | ||
HR+/HER2+ Postmenopausal Locally advanced or MBC No prior systemic treatment | Ribociclib + trastuzumab + letrozole | PFS OS | Recruiting | Phase Ib/II NCT03913234 | ||
HER2+ Locally advanced or MBC Prior systemic treatment | Ribociclib monotherapy | MTD PFS ORR CBR OS | Active, not recruiting | Phase Ib/II NCT02657343 | ||
HER2- Locally advanced or MBC Prior chemotherapy treatment | Ribociclib + capecitabine | MTD Safety Efficacy | Recruiting | Phase I dose-escalation NCT02754011 | ||
TNBC AR+ MBC or unresectable BC Prior systemic treatment | Ribociclib monotherapy | MTD PFS ORR CBR OS | Active, not recruiting | Phase I/II NCT03090165 | ||
Abemaciclib | HR+/HER2- Post-menopausal Stage I to III Prior endocrine treatment | Abemaciclib + fulvestrant | pCR ORR RFS | Recruiting | Phase II NCT04305236 | |
HR+/HER2- Stage II to III No prior systemic treatment | Abemaciclib + letrozole | iEFS CR | Recruiting | Phase II NCT04293393 | ||
HR+/HER2- Locally advanced or MBC Nor prior systemic treatment | Abemaciclib + AIs | ORR CBR TTP DoCB | Recruiting | Phase II NCT04227327 | ||
HER2+ Locally advanced or MBC Prior systemic treatment | Abemaciclib + TDM-1 vs. TDM-1 alone | ORR OS | Recruiting | Phase II NCT04351230 | ||
TNBC Rb+ Locally advanced or MBC Prior chemotherapy treatment | Abemaciclib monotherapy | ORR PFS OS CBR | Recruiting | Phase II NCT03130439 | ||
Palbociclib | HR+/HER2- Post-menopausal Locally advanced or MBC Prior chemotherapy treatment | Palbociclib + fulvestrant | PFS ORR CBR OS | Recruiting | Phase II NCT04318223 | |
ER+ Stage I to III No prior systemic treatment | Palbociclib + endocrine therapy vs. endocrine therapy alone | pCR Safety Tolerability | Recruiting | Phase I NCT03573648 | ||
ER+/HER2+ MBC Prior systemic treatment | Palbociclib + letrozole and TDM-1 | ORR CR SD | Active, not recruiting | Phase I/II NCT03709082 | ||
HER2+ Post-menopausal MBC No prior systemic treatment | Palbociclib + anastrozole + trastuzumab + pertuzumab | DLT MTD CBR PFS | Recruiting | Phase I/II NCT03304080 | ||
HER2+ Rb+ MBC Prior anti-HER2 treatment | Palbociclib + TDM-1 | MTD DLT | Active, not recruiting | Phase Ib NCT01976169 | ||
Antibodies drug conjugates | Trastuzumab-deruxtcan | HER2+ Unresectable or MBC Prior TDM-1 treatment | Trastuzumab-deruxtcan vs. trastuzumab + capecitabine vs. lapatinib + capecitabine | PFS OS ORR DoR | Active, not recruiting | Phase III NCT03523585 |
HER2+ Unresectable or MBC Prior anti-HER2 treatment | Trastuzumab-deruxtcan vs. TDM-1 | PFS OS ORR DoR | Active, not recruiting | Phase III NCT03529110 | ||
HER2- Unresectable or MBC Prior systemic treatment | Trastuzumab-deruxtcan vs. chemotherapy | PFS OS ORR DoR | Active, not recruiting | Phase III NCT03734029 | ||
Trastuzumab-duocarmycin | HER2+ Locally advanced or MBC Prior anti-HER2 treatment | Trastuzumab-duocarmycin vs. standard treatment | PFS OS ORR | Active, not recruiting | Phase III NCT03262935 | |
RC48 | HER2+ Locally advanced or MBC Prior systemic treatment | RC48 vs. lapatinib + capecitabine | PFS ORR DoR CBR OS | Recruiting | Phase II NCT03500380 | |
HER2+ or HER2- Locally advanced or MBC No prior systemic treatment | RC48 monotherapy | ORR CBR PFS | Recruiting | Phase Ib NCT03052634 | ||
PF06804103 | HER2+ or HER2- Solid tumors | PF06804103 alone vs. PF06804103 + letrozole and palbociclib | DLT PFS TTP | Recruiting | Phase I dose-escalation NCT03284723 | |
Ladiratuzumab vedotin | TNBC Locally advanced or MBC No prior chemotherapy | Ladiratuzumab vedotin monotherapy | DLT ORR DoR PFS OS | Recruiting | Phase I NCT01969643 | |
Bispecific antibodies | MCLA-128 | HER2+ or ER+/HER2- Locally advanced or MBC No prior systemic treatment | MCLA-128 + trastuzumab vs. MCLA-128 + trastuzumab and vinorelbine or MCLA-128 + endocrine therapy | CBR PFS ORR DoR OS | Active, not recruiting | Phase II NCT03321981 |
ZW25 (Zanidatamab) | HR+/HER2+ Locally advanced or MBC Prior anti-HER2 treatment | ZW25 + Palbociclib + fulvestrant | DLT PFS IAEs | Recruiting | Phase IIa NCT04224272 | |
ISB 1302 | HER2+ MBC Prior anti-HER2 treatment | ISB 1302 monotherapy | MTD IAEs | Terminated | Phase I/II NCT03983395 | |
PRS-343 | HER2+ solid tumors No prior systemic treatment | PRS-343 + atezolizumab | DLT ORR DoR CR IAEs | Active, not recruiting | Phase Ib NCT03650348 | |
HER2+ solid tumors Locally advanced or MBC | PRS-343 monotherapy | IAEs | Recruiting | Phase I NCT03330561 | ||
Androgen receptor inhibitors | Bicalutamide | TNBC AR+ Locally advanced or MBC | Bicalutamide alone vs. chemotherapy | PFS CBR ORR OS | Terminated | Phase III NCT03055312 |
TNBC AR+ Unresectable or MBC Up to one prior systemic treatment | Bicalutamide + ribociclib | MTD CBR ORR PFS OS | Active, not recruiting | Phase I/II NCT03090165 | ||
TNBC or HER2+ AR+ Stage IV MBC Prior systemic treatment | Bicalutamide monotherapy | pCR PFS Safety | Active, not recruiting | Phase II NCT00468715 | ||
TNBC or ER+ AR+ MBC Prior systemic treatment | Bicalutamide + Palbociclib | PFS CBR Safety Tolerability | Active, not recruiting | Phase I/II NCT02605486 | ||
Enzalutamide | TNBC AR+ Stage I to III No prior treatment | Enzalutamide + paclitaxel | pCR PFS | Recruiting | Phase IIb NCT02689427 | |
TNBC AR+ PTEN+ Stage III to IV MBC No prior treatment | Enzalutamide + alpelisib | MTD PFS CBR | Recruiting | Phase Ib NCT03207529 | ||
TNBC AR+ Stage I to III Prior chemotherapy treatment | Enzalutamide monotherapy | TDR | Active, not recruiting | Feasibility study NCT02750358 | ||
CR1447 | ER+ or TNBC AR+ MBC One prior systemic treatment | CR1447 monotherapy | CR PR SD | Active, not recruiting | Phase II NCT02067741 | |
Anti-PD1 antibodies | Pembrolizumab | HR+/HER2- Locally advanced or MBC Prior chemotherapy and CDK4/6 inhibitors treatments | Pembrolizumab + paclitaxel | ORR CBR PFS DoR OS | Recruiting | Phase II NCT04251169 |
HER2+ MBC Prior systemic treatment No prior TDM-1 treatment | Pembrolizumab + TDM-1 | ORR PFS DoR OS | Active, recruiting | Phase Ib NCT03032107 | ||
HR+/HER2- MBC Prior systemic treatment | Pembrolizumab + fulvestrant | ORR PFS | Recruiting | Phase II NCT03393845 | ||
HR+ or TNBC MBC Prior systemic treatment | Pembrolizumab + Nab-paclitaxel | ORR PFS OS | Recruiting | Phase II NCT02752685 | ||
TNBC Prior systemic treatment | Pembrolizumab + cyclophosphamide | PFS | Active, recruiting | Phase II NCT02768701 | ||
TNBC MBC Prior systemic treatment | Pembrolizumab + Carboplatin and Nab-paclitaxel | PFS DCR | Active, recruiting | Pilot study NCT03121352 | ||
TNBC or ER+ or HER2+ BRCA mutated Locally advanced or MBC Prior systemic treatment | Pembrolizumab + olaparib | ORR PFS OS CBR DoR | Recruiting | Phase II NCT03025035 | ||
Anti-CTLA-4 antibodies | Tremelimumab | HR+/HER2- Stage I to III No prior systemic treatment | Tremelimumab + durvalumab | IAEs pCR | Active, not recruiting | Pilot study NCT03132467 |
HER2-derived vaccines | E75 | HER2+ Stage I to III Prior systemic treatment | E75 vaccine + trastuzumab vs. trastuzumab + GM-CSF | DFS RFS | Active, not recruiting | Phase II NCT02297698 |
GP2 | HER2+ Prior systemic treatment except for trastuzumab | G2P vaccine + GM-SCF and trastuzumab vs. trastuzumab | IAEs | Active, not recruiting | Phase Ib NCT03014076 | |
AE37 | TNBC Prior systemic treament | AE37 vaccine + pembrolizumab | ORR PFS OS CBR | Active, not recruiting | Phase II NCT04024800 | |
Other vaccines | PVX-140 | TNBC HLA-2+ Stage II or III Prior systemic treatment | PVX-140 + durvalumab | DLT DFS IAEs | Active, not recruiting | Phase Ib NCT02826434 |
Neoantigen DNA vaccine | TNBC Post-menopausal Prior systemic treatment | Neoantigen DNA vaccine + durvalumab vs. Neoantigen DNA vaccine alone | Safety Immune response | Recruiting | Phase I NCT03199040 | |
Dendritic cell vaccine | TNBC or ER+/HER2- Locally advanced | DC vaccine + chemotherapy | Safety pCR DFS | Completed | Pilot study NCT02018458 |
TNBC: triple negative breast cancer; HER2: human epidermal growth factor receptor 2; ER: estrogen receptor; MBC: metastatic breast cancer; BC: breast cancer; HR: hormonal receptor; PFS: progression free survival; CBR: clinical benefit rate; ORR: objective response rate; DFS: disease-free survival; OS: overall survival; TTP: time to progression. pCR: pathologic complete response; GM-CSF: granulocyte macrophage colony-stimulated factor; DLT: dose-limiting toxicities; MTD: maximum tolerated dose; TTF: time to treatment failure; TTR: time to treatment response; iDFS: invasive disease-free survival; RFS: recurrence free survival; DDFS: distant disease-free survival; iEFS: invasive events-free survival; CR: clinical response; DoCB: duration of clinical benefit; SD: stable disease; DoR: duration of response; IAEs: incidence of adverse events; TDR: treatment discontinuation rate; PR: partial response; DCR: disease control rate; HR: hazard ratio.
Targeted therapy is the current standard of care to treat HR+ and HER2+ BC, but it cannot be administered to patients with TNBC as these tumors lack the expression of these biomarkers. Hence, the next logical step is to identify biomarkers associated with TNBC to develop specific targeted therapies. Several emerging targeted therapies are being clinically trialed with limited or mixed results.
Vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) are overexpressed in most TNBC patients [ 341 , 342 ]. Bevacizumab and cetuximab are antibodies developed to specifically target VEGF and EGFR, respectively. Unfortunately, clinical trials studying the effects of these antibodies in TNBC patients demonstrated limited results. The phase III, randomized BEATRICE study evaluating adjuvant bevacizumab-continuing therapy in TNBC demonstrated no significant benefit in OS [ 310 ]. A phase II trial evaluating the impact of adding bevacizumab or cisplatin to neoadjuvant chemotherapy to stage II to III TNBC concluded that further investigation of bevacizumab in this setting was unlikely [ 311 ].
The phase II randomized TBCRC 001 trial testing the combination of cetuximab and carboplatin in stage IV TNBC showed a response in fewer than 20% of patients [ 312 ]. Another randomized phase II study compared the effects of cetuximab plus cisplatin to cisplatin alone in metastatic TNBC patients. Adding cetuximab to cisplatin prolonged PFS and OS, warranting further investigation of cetuximab in TNBC [ 313 ]. Based on these results, bevacizumab is not recommended for the treatment of TNBC.
mTOR/PI3K/Akt signaling pathway is an important target involving all BC subtypes. Inhibitors of mTOR, PI3K, and Akt have been tested in HR+ and HER2+ BC patients and have also been tested in TNBC patients. The mTOR inhibitor everolimus has been tested in a randomized phase II trial in combination with chemotherapy vs. chemotherapy alone in stage II/III TNBC patients [ 314 ]. Unfortunately, the addition of everolimus was associated with more adverse effects, without improving pCR or clinical response. A phase I study testing the combination of everolimus and eribulin in metastatic TNBC patients showed that this combination was safe, but the efficacy was modest [ 343 ].
The Akt inhibitor ipatasertib has been tested in combination with paclitaxel (vs. placebo) for metastatic TNBC patients in the phase II multicentered double-blinded randomized LOTUS trial [ 315 ]. The results showed improved PFS when patients received ipatasertib. Another phase II double-blinded randomized trial, FAIRLANE, testing neoadjuvant ipatasertib plus paclitaxel for early TNBC, showed no clinically or statistically significant improvement in the pCR rate, but ipatasertib’s antitumor effect was more pronounced in patients with PI3K/AKT1/PTEN-altered tumors [ 316 ]. Capivasertib, another Akt inhibitor, has been tested in combination with paclitaxel (vs. placebo), first-line therapy for metastatic TNBC patients in the phase II double-blinded randomized PAKT trial [ 317 ]. The addition of capivasertib to paclitaxel significantly improved PFS and OS, with better benefits for patients with PI3K/AKT1/PTEN-altered tumors.
The androgen receptor (AR) is a steroidal hormonal receptor that belongs to the nuclear receptor family and is expressed in 10% to 50% of TNBC tumors [ 344 ]. Tumors expressing AR have better prognosis but are less responsive to chemotherapy [ 345 ]. Multiple clinical trials have tested AR inhibitors in TNBC [ 318 , 319 , 320 ].
Bicalutamide, an AR agonist, was tested in a phase II study in patients with AR+, HR- metastatic BC [ 318 ]. The results showed promising efficacy and safety for these patients.
Enzalutamide, a nonsteroidal antiandrogen, has been tested in a phase II study in patients with locally advanced or metastatic AR+ TNBC [ 319 ]. Enzalutamide demonstrated significant clinical activity and tolerability, warranting further investigation.
Abiraterone, a selective inhibitor of CYP17, has been evaluated in combination with prednisone in AR+ locally advanced or metastatic TNBC patients [ 320 ]. This combination was beneficial for 20% of the patients.
Several clinical trials are currently testing AR inhibitors alone or combined with other treatments for TNBC patients; expecting results between 2022 and 2027 (see Table 4 ).
Targeted antibodies.
The immune system plays a crucial role in BC development and progression. Tumor cells can escape the immune system by regulating T-cell activity leading to the inhibition of immune response [ 346 , 347 ]. Two principal biomarkers found in TNBC are associated with this bypass: the programmed cell death protein receptor (PD-1) and its ligand PDL-1, and the cytotoxic T lymphocyte-associated protein 4 (CTLA-4) [ 348 ].
PD-1 is an immune checkpoint receptor expressed on the surface of activated T-cells. PDL-1, its ligand, is expressed on the surface of dendritic cells or macrophages. The interaction of PD-1 and PDL-1 inhibits T-cell response [ 349 ]. CTLA-4 is expressed on T-cells and inhibits T-cell activation by binding to CD80/CD86, leading to decreased immune response [ 350 ].
Atezolizumab, an anti-PDL-1 antibody, has demonstrated safety and efficacy in a phase I study for metastatic TNBC patients [ 351 ]. Based on these results, atezolizumab was tested in combination with nab-paclitaxel for unresectable locally advanced or metastatic TNBC in the phase III double-blinded placebo-controlled randomized Impassion130 study [ 321 ]. Atezolizumab plus nab-paclitaxel prolonged PFS and OS in both the intention-to-treat population and PDL1+ subgroup. Another double-blinded, randomized phase III study (Impassion031) compared atezolizumab in combination with nab-paclitaxel and anthracycline-based chemotherapy vs. placebo for early-stage TNBC [ 322 ]. This combination significantly improved pCR with an acceptable safety profile.
Durvalumab, another anti-PDL-1 antibody, has been tested in combination with an anthracycline taxane-based neoadjuvant therapy for early TNBC in the randomized phase II GeparNuevo study [ 323 ]. This combination increased pCR rate, particularly in patients pretreated with durvalumab monotherapy before chemotherapy. Another randomized phase II study, SAFIRO BREAST-IMMUNO, compared durvalumab to maintenance chemotherapy in a cohort including TNBC patients [ 324 ]. Results showed that durvalumab, as a single agent therapy, could improve outcomes in TNBC patients. A phase I study tested durvalumab in combination with multiple TNBC therapies: PARP inhibitor olaparib and VEGFR1-3 inhibitor cediranib for patients with recurrent cancers including TNBC [ 325 ]. This combination was well tolerated and showed preliminary antitumor activity in all of these patients.
The safety and efficacy of avelumab, another anti-PDL-1 antibody, was evaluated in the phase Ib JAVELIN study in patients with locally advanced or metastatic BC, including TNBC [ 326 ]. Avelumab showed an acceptable safety profile and clinical activity, particularly in tumors expressing PDL-1.
Pembrolizumab is an anti-PD-1 antibody that has been tested in multiple clinical trials. The phase Ib KEYNOTE-012 study demonstrated the safety and efficacy of pembrolizumab on advanced TNBC patients [ 352 ]. Based on these results, the phase II KEYNOTE-086 study evaluated pembrolizumab monotherapy for pretreated or non-pretreated metastatic TNBC patients [ 327 , 353 ]. Pembrolizumab monotherapy showed a manageable safety profile and durable antitumor activity for both pretreated and non-pretreated subgroups. The randomized open-labeled phase III KEYNOTE-119 trial compared pembrolizumab monotherapy to standard chemotherapy in metastatic TNBC [ 354 ]. Pembrolizumab monotherapy did not significantly improve OS compared to chemotherapy in these patients. These findings suggest that pembrolizumab should be investigated in a combinational approach rather than in monotherapy. Based on these results, pembrolizumab was tested in combination with chemotherapy (vs. placebo) for pretreated locally recurrent or metastatic TNBC patients in the phase III double-blinded randomized KEYNOTE-355 trial [ 328 ]. The combination of pembrolizumab plus chemotherapy significantly and clinically improved PFS compared to chemotherapy plus placebo. Pembrolizumab has also been evaluated for early TNBC as neoadjuvant therapy in combination with chemotherapy (vs. placebo) in the phase III KEYNOTE-522 trial [ 329 ]. The combination of pembrolizumab plus chemotherapy significantly improved pCR rate in these patients compared to placebo plus chemotherapy.
Tremelimumab is an anti-CTLA-4 antibody. A dose-escalation phase I study evaluating the safety and efficacy of tremelimumab in patients with metastatic BC showed good tolerability [ 330 ].
Vaccination is an emerging approach to prevent recurrence in high-risk BC patients. As mentioned earlier, TNBC is the most aggressive BC subtype with a higher risk of distant recurrence [ 331 ]. Thus, developing vaccines to prevent recurrence in TNBC patients is of great interest.
Takahashi et al. have developed a novel regimen of personalized peptide vaccination (PPV) based on the patient’s immune system to select vaccine antigens from a pool of peptide candidates [ 332 ]. They performed a phase II study where metastatic recurrent BC patients with prior chemotherapy and/or hormonal therapies received a series of personalized vaccines. This vaccination demonstrated safety, possible clinical benefit, and immune response, especially for TNBC patients [ 332 ]. A multicentered, randomized, double-blinded phase III study analyzed the effects of sialyl-TN keyhole limpet hemocyanin (STn-KLH) on metastatic BC patients [ 333 ]. STn-KLH consists of a synthetic STn, an epitope expressed in BC and associated with aggressive and metastatic tumors, and a high molecular weight protein carrier KLH [ 355 ]. Stn-KLH demonstrated good tolerability, but no benefits in time to progression (TTP) or survival were found. Thus, this vaccination is not recommended for metastatic BC patients [ 333 ].
PVX-410 is a multiple peptide vaccine that activates T-cell to target tumor cells and was developed to treat myeloma. A phase Ib/II study demonstrated the safety and immunogenicity in myeloma patients [ 356 ]. Based on these results, a PVX-410 vaccine is currently being tested to treat TNBC in multiple clinical trials (see Table 4 ).
Finding new treatments for TNBC is an ongoing challenge. The therapeutic strategies that have been described in this section offer great hope to treat TNBC patients. However, because TNBC is highly heterogeneous, it is difficult to find a single treatment efficient for all TNBC subtypes [ 228 ].
This review clearly demonstrates that the treatment of BC is complex and is constantly evolving with a large number of ongoing clinical trials on emerging therapies. Indeed, the BC molecular subtype will determine the personalized therapeutic approach, such as targeted treatments like endocrine therapy for HR+ BC or anti-HER2 therapy for HER2+ BC. These therapies have demonstrated their safety and efficacy in treating BC over the years. However, it is essential to go beyond these conventional treatments as BC is a complex disease and not all patients can benefit from personalized treatment. One of the major challenges in BC treatment is finding effective therapies to treat TNBC patients since conventional targeted therapies cannot be administered for this specific BC subtype, which has the worst survival outcomes.
Another important issue in BC treatment is the acquisition of treatment resistance. This is a common phenomenon for either endocrine therapy, anti-HER2 therapy, and chemotherapy.
Hence, understanding the mechanisms underlying drug resistance is a good strategy to develop novel treatments for BC. For example, the mTOR/PI3K/Akt pathway is involved in the mechanism of resistance in all BC molecular subtypes, and thus developing specific inhibitors targeting this pathway is a promising BC treatment approach.
The authors would also like to thank team members from the C.D. and F.D. research groups for their valuable assistance.
ABC | ATP binding cassette |
ADC | antibody-drug conjugate |
ADCC | antibody dependent cell cytotoxicity |
AI | aromatase inhibitor |
AIB1 | amplified in breast cancer 1 |
ALND | axillary lymph node dissection |
AR | androgen receptor |
ATM | ataxia-telangiesctasia mutated |
BC | breast cancer |
BCRP | breast cancer resistant protein |
BRCA | breast cancer gene |
BsAb | bispecific antibody |
CBR | clinical benefice rate |
CDK4/6 | cyclin-dependent kinase |
CR | clinical response |
CSC | cancer stem cell |
CTLA4 | cytotoxic T lymphocyte-associated protein 4 |
DDFS | distant disease-free survival |
DFS | disease-free survival |
DLT | dose-limiting toxicities |
DoCB | duration of clinical benefit |
DoR | duration of response |
EGF | epidermal growth factor |
EGFR | epidermal growth factor receptor |
ER | estrogen receptor |
FDA | food and drug administration |
gBRCAm | germline BRCA mutation |
HB-EGF | heparin-binding EGF-like growth factor |
HER2 | human epidermal growth factor receptor 2 |
HGF | hepatocyte growth factor |
HIF1-α | hypoxia-inducible factor 1 alpha |
HR | hormone receptor |
HR | hazard ratio |
IAES | incidence of adverse events |
IDFS | invasive disease-free survival |
iEFS | invasive events-free survival |
IGF-1 | insulin growth factor 1 |
IGF-1R | insulin growth factor receptor 1 |
MAP | microtubule associated protein |
MAPK | mitogen activated protein kinase |
MBC | metastatic breast cancer |
MTD | maximum tolerated dose |
mTOR | mammalian target of rapamycin |
NAC | neoadjuvant chemotherapy |
ORR | overall response rate |
OS | overall survival |
PARP | poly-(ADP-ribose) polymerase protein |
PARPi | poly-(ADP-ribose) polymerase protein inhibitor |
pCR | predicted complete response |
PD-1 | programmed cell death protein receptor |
PDL-1 | programmed cell death protein ligand |
PFS | progression-free survival |
PI3K | phosphoinositide 3-kinase |
PPV | personalized peptide vaccine |
PR | progesterone receptor |
PR | partial response |
PTEN | phosphatase and tensin homolog |
Ras-ERK | extracellular-signal-regulated kinase |
RFS | recurrence-free survival |
SD | stable disease |
SERD | selective estrogen receptor degrader |
SERM | selective estrogen receptor modulator |
SLNB | sentinel lymph mode biopsy |
STnKLH | sialyl-TN keyhole limpet hemocyanin |
T-DM1 | trastuzumab-emtansine |
TKI | tyrosine kinase inhibitor |
TNBC | triple-negative breast cancer |
Trop2 | trophoblast antigen 2 |
TTF | time to treatment failure |
TTP | time to treatment progression |
TTR | time to treatment response |
VEGF | vascular endothelial growth factor |
A.B. conceptualized and drafted the manuscript. F.D. and C.D. supervised the project. All authors did critical revision of the manuscript. All authors have read and agreed to the published version of the manuscript.
This work was supported by the “Fond de recherche du Québec–Santé (FRQS)” associated with the Canadian Tumor Repository Network (CTRNet). Caroline Diorio is a senior Research Scholar from the FRSQ. Anna Burguin holds a Bourse d’excellence en recherche sur le cancer du sein—Faculté de médecine-Université Laval.
Not applicable.
Data availability statement, conflicts of interest.
The authors declare no conflict of interest.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Breast Cancer Research is presenting our Retrospective Collection on "Breast Cancer Risk Factors." Celebrating 'Breast Cancer Awareness Month (1 October- 31 October)', with this Collection, we aim to gain valuable insights into the multifaceted aspects of breast cancer risk to promote awareness, prevention, and early detection.
NEW CROSS-JOURNAL COLLECTIONS Find out more by clicking the links below:
Artif icial Intelligence in Breast Imaging PDGFB in Br east Cancer Initiation,Progression, and Metastasis
Authors: Hui-Chen Wu, Yunjia Lai, Yuyan Liao, Maya Deyssenroth, Gary W. Miller, Regina M. Santella and Mary Beth Terry
Authors: Guilherme Nader-Marta, Christian Singer, Dominik Hlauschek, Angela DeMichele, Paolo Tarantino, Evandro de Azambuja, Georg Pfeiler, Miguel Martin, Justin M. Balko, Zbigniew Nowecki, Marija Balic, Adam M. Brufsky, Arlene Chan, Patrick G. Morris, Tufia Haddad, Sibylle Loibl…
Authors: Jessica O’Driscoll, Anya Burton, Gertraud Maskarinec, Beatriz Perez-Gomez, Celine Vachon, Hui Miao, Martín Lajous, Ruy López-Ridaura, A. Heather Eliassen, Ana Pereira, Maria Luisa Garmendia, Rulla M. Tamimi, Kimberly Bertrand, Ava Kwong, Giske Ursin, Eunjung Lee…
Authors: Bruno Valentin Sinn, Katharina Sychra, Michael Untch, Thomas Karn, Marion van Mackelenbergh, Jens Huober, Wolfgang Schmitt, Frederik Marmé, Christian Schem, Christine Solbach, Elmar Stickeler, Hans Tesch, Peter A. Fasching, Andreas Schneeweiss, Volkmar Müller, Johannes Holtschmidt…
Authors: Bitao Jiang, Lingling Bao, Songqin He, Xiao Chen, Zhihui Jin and Yingquan Ye
Most recent articles RSS
View all articles
Authors: Nusayba Bagegni, Shana Thomas, Ning Liu, Jingqin Luo, Jeremy Hoog, Donald W. Northfelt, Matthew P. Goetz, Andres Forero, Mattias Bergqvist, Jakob Karen, Magnus Neumüller, Edward M. Suh, Zhanfang Guo, Kiran Vij, Souzan Sanati, Matthew Ellis…
Authors: Deborah L Holliday and Valerie Speirs
Authors: Li Yin, Jiang-Jie Duan, Xiu-Wu Bian and Shi-cang Yu
Authors: Diane Scutt, Gillian A Lancaster and John T Manning
Authors: Suzanne A Eccles, Eric O Aboagye, Simak Ali, Annie S Anderson, Jo Armes, Fedor Berditchevski, Jeremy P Blaydes, Keith Brennan, Nicola J Brown, Helen E Bryant, Nigel J Bundred, Joy M Burchell, Anna M Campbell, Jason S Carroll, Robert B Clarke, Charlotte E Coles…
Most accessed articles RSS
Lewis Chodosh , University of Pennsylvania, USA
Click here to see the most popular articles published in Breast Cancer Research in the past three months.
20 years ago Breast Cancer Research published its first articles with BMC. Well-respected in the field, the journal has continually placed in the first quartile of the ‘Oncology’ category of Journal Citation Reports. Over the past decade, Breast Cancer Research (BCR) has also become the highest ranked breast cancer focused title in the field.
Look back at the journal’s milestone achievements and article highlights .
In this review, we provide a useful reference for AI researchers investigating image-based breast cancer risk assessment while indicating key priorities and challenges that, if properly addressed, could accelerate the implementation of AI-assisted risk stratification to future refine and individualize breast cancer screening strategies.
Discover the range of academic oncology titles at Springer Nature here .
Introduction.
Breast cancer is the most frequently diagnosed cancer with one in eight woman being diagnosed in their lifetime 1 . Invasive breast cancers exhibit different histological subtypes with the majority (~75–80%) being carcinoma of no special type (NST), previously referred to as invasive ductal carcinoma (IDC). Invasive lobular breast cancer (ILC) is the most common special type, representing 10–15% of all breast cancers 2 . The hallmark of ILC is loss of E-cadherin, resulting in discohesive cells and alteration of tumor cellular morphology. These are the foundation for the histopathological diagnosis 3 . The diagnosis of ILC remains challenging in part due to the existence of different ILC subtypes and a lack of consistency of diagnostic methodology 4 . Recent studies have shown that interobserver agreement in diagnosis of ILC can be increased with the use of E-cadherin immunohistochemistry (IHC) as a diagnostic marker 5 , and although efforts to improve this have recently been published ( https://pubmed.ncbi.nlm.nih.gov/38641322/ ), at this point in time there are no guidelines recommending its use. The pathognomonic feature of ILC is genetic inactivation of CDH1 6 . Compared to patients with NST, patients with ILC are older, and at the time of diagnosis tumors are mostly estrogen receptor-positive, larger, and of higher stage, likely due to limitations of imaging modalities for detection of lobular tumors 7 . Patients with ILC suffer more frequently from late recurrences (often to less common sites such as the gastrointestinal and urogenital tract) than patients with NST, resulting in worse long-term outcome despite fewer high-risk patients being identified by molecular profiling 8 . There are limited studies comparing response to chemotherapy, but collectively they suggest decreased efficacy in patients with ILC 9 , 10 . Despite the increasing realization of unique biology, etiology, and progression of ILC, ILC is understudied relative to other breast cancers, and there are no treatment guidelines specifically for ILC 7 , however some clinical trials specific for or enriching for patients with ILC are currently being performed.
Here we undertook a worldwide survey, which included the three major stakeholders – breast cancer clinicians/researchers, laboratory-based researchers, and the community including patients and patient advocates. Using this survey, we analyzed the current understanding of ILC and identified consensus research questions on ILC, which can provide the foundation for future collaborative studies.
In total, 1714 individuals were contacted by email with a 39% response rate and for these responses there was a 95% completion rate in taking the survey. Subsequent additional targeted recruitment and outreach via social media especially Twitter (now X) and Facebook resulted in an additional 1,166 respondents starting the survey. In total, 1774 participants answered at least one question and 1310 finished the survey. Of the survey respondents, 688 asked to be included as having contributed to the survey, and they are listed (Supplementary Data File 1 ).
Respondents resided in 66 countries (Supplementary Data File 2 ) covering all continents except Antarctica (Fig. 1A ). The top three countries based on number of respondents were the US (47.65%), UK (9.66%) and Ireland (6.55%).
A A world map showing the percentage by country of respondents to the survey, and the top ten countries with numbers of respondents. World map was generated using https://github.com/geopandas/geopandas . B Overall number of physicians, lab- based researchers, and breast cancer patients, with advocates being indicated. C Physicians are separated by clinical specialty.
Respondents self-identified as breast cancer physicians/researchers ( N = 413), as basic (laboratory-based) researchers ( N = 376), and breast cancer patients ( N = 1121) of whom 288 (26.1%) indicated membership in advocacy groups (Fig. 1B ). Some respondents belonged to more than one category - 28 clinicians and 17 laboratory-based researchers were also breast cancer patients. There was also overlap between the breast cancer clinicians/researchers and the laboratory-based researcher, with 195 (47.2%) identifying as clinicians with a research lab.
The majority of the breast cancer physicians were medical oncologists (46.5%) and surgeons (22.5%) followed by representation from pathology (12.3%), radiation oncology (4.1%), gynecology (1.0%) and others (5.6%), which included palliative care, patient navigators, clinical oncology, geneticist, family medicine, nuclear medicine and more (Fig. 1C ). Most of the clinicians practice in academia (60.6%), followed by private practice (17.4%), and governmental institutions (16.9%). There was an equal distribution of experience: 1–10 years (24.0%), 11–20 years (32.6%), 21–30 years 22.7%), and 31 years and more (19.6%). The majority of physicians treated 11-50 (42.8%) and 51–100 (26.8%) patients per month.
The majority of self-identified laboratory-based researchers work in academic institutions (80.7%), and others in private (9.4%) and governmental (6.7%) institutions. There was a roughly equal distribution of researchers working (50.9%) vs. not working on lobular (49.1%) breast cancer, and 80% of those working on ILC have previously received funding for their work on ILC from a wide range of funders (Supplementary Data File 3 ). The majority of researchers have been working for 1–10 years (35.7%), followed by 11–20 years (29.7%), 21–30 years (23.7%), 31 years (7.8%) and more (19.6%), and less than 1 year (3.6%).
There were 1121 respondents that indicated that they have or have had breast cancer or in situ carcinoma, called “patients” hence forth. At the time of response to the survey, 62.5% had breast cancer but were currently without evidence of disease, 25.4% were in active treatment for breast cancer, and 19.1% had indicated that their disease had recurred with 8.1% being local and 11.1% being distant recurrences. Lobular breast cancer was the most common histology (63.1%), followed by LCIS (13.6%), DCIS (6.8%), NST (6.3%), mixed ductal/lobular (5.3%) and others/unknown (4.9%). The average age for diagnosis was 52.3 years.
Of the 1121 patients who responded, 288 (26.1%) indicated membership in advocacy groups. Of the total number of respondents to the survey, 403 (23.1% of all respondents) belonged to one or more of 170 different advocacy groups, support groups, and other foundations (Supplementary Data File 4 ).
The majority of physicians were confident in describing the differences between ILC and NST (Fig. 2A ), and there was no significant difference between medical oncologists, surgeons and other specialties. Only 4% were not all confident, or slightly confident (12%) about the differences. Physicians indicated that knowledge of histology was seen as very/extremely important (73%) (Fig. 2B ), and this again was not different between the different specialties The majority of physicians stated that knowledge of histology affected their treatment decisions a lot (51%) or a moderate amount (32%), with surgeons (60%) and others (59%) using histology information significantly more than medical oncologists (40%). Very few physicians (4.5%) felt that knowing histology was not at all or only slightly important. The majority of physicians (57%) indicated that there either was no data, that they did not know, or that they were unsure if there were clinical trials and outcome data supporting unique treatments for ILC. Refining treatment guidelines specifically for lobular breast cancer was seen as valuable for treating patients with ILC in the future by 76% of physicians, for a number of reasons outlined in Supplementary Text File 2 .
A Responses by clinicians about confidence in describing differences between ILC and NST. B Importance of knowledge of histology for physicians. C Patients’ responses on communication with physicians. ”Other” refers to “ I was not offered personalized therapy because my physician explained that the treatment is no different for ductal than for lobular ”.
We asked how patients perceived the knowledge of and specifically, the communication about ILC-specific features by physicians. Many patients (52%) thought that their health care providers did not explain unique features of ILC (Fig. 2C ). “Personalized therapy based on histological diagnosis” was discussed with 21% of patients, while it was not discussed with 42%. For 28% of patients, the physicians explained that there was no difference in treatment for NST and ILC. Discussions about potential personalized therapies were held mostly with medical oncologists (89%), followed by surgeons (72%). There were an equal number of radiation oncologists who “definitely did not/probably not” (50%) and who “definitely did/probably did” (50%) discuss ILC-personalized therapy. Most physicians (71%) did not mention that ILC can metastasize to unique places and did not discuss what symptoms, including unusual symptoms, of recurrence the patients should report in the future (78%). For all these communication-related questions, there were differences between countries, with health care providers in the US more frequently being perceived as explaining ILC specific features compared to other countries (Supplementary Data File 5 ).
Finally, we asked which other ILC-related topics the patients wished they have had a chance to discuss with their physicians. The most common was a discussion of the unique clinical features of ILC, followed by information on recurrence and metastasis, cancer detection and screening, and influence of breast density (Supplementary Data File 6 , and Supplementary Fig. 1 ). All answers were clustered into one of the topics based on semantic similarity, which can be interactively visualized under ‘Topic’ coloring scheme via https://atlas.nomic.ai/data/chelseax488/ilc-survey---discussion-with-doctors/map .
For those who self-identified as basic or translational researchers, 48% were very/extremely and 29% were moderately confident in describing differences between ILC and NST. The majority (59.8%) performed none or only a little ILC research with only 20% performing a lot or a great deal of ILC research. Reflecting this, only 23% received funding to work on ILC, and those who focused on ILC were significantly more funded for their ILC work (54%) compared to those who don’t focus on ILC (9%).
There is the need for additional ILC models, as only 11% of respondents found that there were sufficient in vitro and in vivo models for ILC research. 32% of respondents use ILC cell line models in their research, the most common being MDA-MB-134 and SUM44. Majority did not use ILC models due to “lack of facility, resources, or expertise”. 52% of respondents felt that ILC was poorly represented in public genomic datasets, while 69% felt that they were able to obtain lobular breast cancer tissue and/or blood samples from patients with ILC for research.
We asked the three major stakeholders for their opinions on research priorities in ILC (Fig. 3 , and Supplementary Data File 7 ). We posed questions about 6 major areas with each area having subcategories. The main areas were: (1) Epidemiology and Risk Reduction; (2) Diagnosis (Imaging and Pathologic Analysis), (3) Therapy, treatment resistance and disease progression; (4) Local therapy of the primary tumor; (5) Imaging; and, (6) Lobular tumorigenesis (the formation of tumors), and other basic/translational research questions. There was general agreement in prioritization of research priorities by the physicians and laboratory-based researchers. Both chose “Therapy, treatment resistance and disease progression” as their top research area followed by “Diagnosis (Imaging and Pathology)”. For 5 out of the 6 areas there was agreement on the specific subcategories of interest with the two highest being “Determining mechanisms of endocrine resistance in ILC” and “Understanding value of genomic predictors for ILC prognosis and prediction of therapeutic response”. The largest discordance was in the area of “Basic/translational research” with physicians choosing “Focusing on development of a centralized ILC data and tissue registry” whereas laboratory-based researchers chose “Characterizing differences in the tumor microenvironment between ILC and NST”. The top priority areas for patients were “Imaging” and “Diagnosis (Imaging and Pathology)”. Within “Imaging”, all groups identified “Identifying new and specific imaging tools for ILC” as the key research area, whereas in “Diagnosis (Imaging and Pathologic Analysis)”, patients chose “Identifying strategies to improve ILC screening/early detection” and physicians and researchers choose “Role of genomic predictors for ILC prognosis and prediction of therapeutic response”.
Heatmap showing percentage of individuals rating each research question as of highest importance (‘most critical and impactful’, against moderate/low importance) among 6 domains of topics in physicians, lab-based researchers, and patients, respectively. Color and number represents the percentage from 0-100 in each block.
And finally, there was a free text field question asking which other research questions have high priority that might not have been listed. The most frequently mentioned topics (Supplementary Data File 8 ) among a wide range of answers were: (1) Genetic screening, Germline mutations, Familial risks, (2) Awareness education, (3) New Aromatase Inhibitors (AI) and Selective Estrogen Receptor Degraders (SERDs) and duration of treatment, (4) Genomic predictors/markers, and, (5) Chemotherapy-related questions.
Finally, we asked physicians specifically about their opinions on clinical trials in ILC. Half of the physicians reported that “Most of the time/always” approximately half of the clinical trials and studies they were involved in collected data on tumor histology, with twice as many surgeons (35%) than medical oncologists (16%) “always” collecting histology information. Further, 66% of physicians stated that clinical trials they are involved in do not or only sometimes consider histology in their inclusion/exclusion criteria. Most physicians (86%) have not powered clinical trials they were involved in to specifically allow a subset analysis for ILC, and this was not significantly different between surgeons, medical oncologists and other physicians. However, 50% would “probably” and 36% would “definitely” consider powering clinical trials to do subset analysis of lobular breast cancer in the future, for a wide range of reasons (Supplementary Text File 3 ). Importantly, most physicians (87%) would consider participating in consortia conducting clinical trials in ILC.
ILC is inherently understudied compared to the more common NST subtype of breast cancer. To better understand conceptions about ILC and key research areas, we surveyed patients/advocates, physicians, and laboratory-based researchers. A robust world-wide response ( n = 1774) was obtained from all three key stakeholder groups. Physicians are confident in describing the differences between ILC and NST and feel that knowledge of histology is important, as it affects their decision making. Importantly, they responded that future refined treatment guidelines would be valuable for patients with ILC. While very few clinical trials have been directed specifically at ILC, most physicians expressed an interest in such trials. Most laboratory researchers felt that ILCs are inadequately presented in large genomic data sets, and there are too few models of the disease. The majority of patients thought that their health care providers did not explain unique features of ILC, and that in general communication was limited. Overall, while there is growing interest in the study and understanding of ILC, there are clear gaps in understanding and presentation of the disease to patients. This challenge is beginning to be addressed by advocacy groups who are developing publicly available educational materials available on websites for examples from the Lobular Beast Cancer Alliance ( https://lobularbreastcancer.org/ ), the European Lobular Breast Cancer Consortium ( https://elbcc.org/ ), and in general there has been an overall increased awareness of unique features of ILC.
Both clinical and laboratory studies are hindered by a lack of studies targeted specifically at ILC and addressing ILC-specific challenges. We asked the three major stakeholders what they felt were the most important areas of ILC research. While there was general concordance between the groups, important areas did show discordance. For example, while physicians and researchers both chose ‘Therapy, treatment resistance and disease progression’ as their top research area, patients/advocates chose ‘Imaging’ and ‘Diagnosis (Imaging and Pathologic Analysis)’. Our survey results are consistent with a recent survey of patients and advocates ( n = 1476) previously diagnosed with ILC who expressed concerns over current imaging standards 11 . These patients reported that mammography often failed to detect ILC cases until they reach stage 2 or higher, a common issue for ILC. Importantly, tumors were often larger at resection than predicted by imaging. A recent review of the literature also reported that MRI and contrast-enhanced mammogram surpass conventional breast imaging in sensitivity and specificity of ILC detection 12 . There have been recent advances in understanding the reasons for decreased ability to image ILC e.g., reduced uptake of glucose limited FDG-PET 13 and direct comparisons showing better imaging via FES-PET compared to FDG-PET in ILC 14 . This is clearly an area of patient/advocate interest.
Patients and advocates also highlighted the need for improved diagnosis (imaging and pathologic analysis). This is also consistent with a recent survey of pathologists highlighting the lack of standard definitions for diagnosis of ILC 4 . The current World Health Organization (WHO) classification for ILC diagnosis only requires pathologists to note a non-cohesive growth pattern in the tumor and does not require analysis of E-cadherin status. Highlighting challenges in pathologic diagnosis of ILC, thirty-five pathologists diagnosed NST and ILC from a set of breast cancers and showed only a moderate inter-observer agreement, but a substantial agreement was a found when E-cadherin was also used in the diagnosis 5 . Efforts have recently been done by the pathology working group from the European Lobular Breast Cancer Consortium (ELBCC) to harmonize pathological diagnosis of ILC ( https://pubmed.ncbi.nlm.nih.gov/38641322/ ). Finally, the increased development and use of digital pathology and artificial intelligence, such a recently reported algorithm using 51 different types of clinical and morphological features differentiating between NST and ILC with an AUC of 0.97 15 may eventually offer the option to improve diagnosis. Another example is a recent study which used a machine learning system to detect ‘ CDH1 biallelic mutations’ as ground truth rather than histology and then developed an AI-based system that can detect ILCs accurately 16 . These algorithms may help resolve the morpho-molecular diagnosis of ILC 17 , particularly with the complex mixed subtypes.
Different from the patients/advocates, the top two most important research questions identified by clinician and laboratory researchers were 1) determining mechanisms of endocrine resistance, and 2) identifying novel therapeutic targets, repurposing existing drugs and progressing them to clinical trials. These research questions align with the identified priority areas of ELBCC ( https://lobsterpot.eu/organisation/working-groups and https://lobsterpot.eu/organisation/pr2 ), and likely reflect the desire to personalize therapy for patients with ILC through improved understanding of unique biology of ER and other signaling pathways. While preclinical and clinical studies have suggested that ILC may be relatively resistant to antiestrogen therapy 18 , 19 , 20 and sensitive to inhibitors of growth factor receptor/PI3K/ROS1 signals 21 , 22 , 23 , 24 , 25 , clinical trial evidence supporting these concepts remain lacking. The first clinical trial that focused specifically on ILC (GELATO; NCT03147040) which evaluated chemotherapy followed by immunotherapy in patients with metastatic ILC showed a relatively low (27%) clinical benefit rate 26 . It was discontinued in part because the responding tumors were of the rare ER-negative subtype of ILC and because chemotherapy plus immunotherapy is now standard of care for patients with PD-L1+ metastatic TNBC, regardless of histological subtype. Of note, interesting correlative science from this trial as well as another recent study 27 supports the need for further analysis of the immune infiltrate in ILC.
There are limitations to the study, which include uneven representation of the three stakeholder groups. In addition, the physicians are not truly a representative group as only 17% are in private practice suggesting limited participation from community physicians. In addition, physicians involved in the diagnostic practices, including pathologists and radiologists, are underrepresented in the survey respondents, which are skewed towards medical oncologists. As with many surveys, the responses are biased in terms of who takes the time to respond to the survey, ie the respondents are likely highly motivated to improve understanding of ILC. Finally, although we do have representation from 66 countries, almost two thirds of the responses (64%) originate from only three countries.
In summary, we have obtained data on world-wide understanding and interest in the study of ILC. Patients feel that communication of the unique features of ILC by the physician can be improved. While physicians and laboratory researchers feel the need to better understand endocrine resistance, to identify new treatment targets and/or repurpose existing drugs for ILC treatment, patients’ top priority research area is improved imaging. Overall, the survey indicates areas where interventions can be implemented to improve communication and outcomes for patients with ILC.
After discussing the concept and development of an early version of the survey, a draft was shared with representatives from the three groups, breast cancer clinicians/researchers, laboratory-based researchers, and the community including patients and patient/advocates (hitherto called “physicians,” “researchers,” and “patients/advocates”). Multiple rounds of edits were circulated among the team, and the survey was then further refined by the UPMC Hillman Cancer Center Population Survey Facility. After beta testing with a subset of physicians, researchers and patients/advocates, final edits were made, and the survey was developed and distributed using the Qualtrics online survey tool, through the Population Survey Facility. The final survey (Supplementary Text File 1 ) was fielded March to May 2022 to email lists compiled from PubMed, ORCID and professional networks, followed by distribution via social media.
For our study, we have complied with all relevant ethical regulations including the Declaration of Helsinki. The survey was anonymized, and responses were not linked to identifiers, however respondents had the opportunity to list names if they chose to be identified as survey participants in the resulting manuscript (Supplementary Data File 1 ). The study received Pitt institutional review board approval (STUDY21010058, initially effective on 03/22/2021, and final version was effective on 02/17/2022).
All responses were collected by the University of Pittsburgh Population Survey Facility. To analyze responses for topic of most critical and impactful research questions, each variable was dichotomized (most impactful or not), a top score was generated which was then analyzed using chi-square analysis across the subgroups.
Free text field questions 34 (“Which ILC cell models are you including in your studies”) and 58 (“Please indicate below other research questions that have high priority that we have not listed”) were manually summarized (SO, AVL) with common themes assigned for each answer with high consistency (overlap rate for Q58 = 98.36%). Natural language processing (Chat GPT, and sentence transformer – see below for details) was used to analyze responses to free text field questions 33 (“Why are you not including ILC cell models in your studies”) and 50 (“What do you now know about ILC that you wish you could have heard from and discussed with your…”), which were all initially filtered to exclude missing or semantically non-meaningful items. For question 33, a prompt was made for ChatGPT4, “Think as a physician scientist, patient, and basic researcher in breast cancer. Summarize the common but non-overlapping reasons shared among answers, ‘Why are you not including ILC cell models in your studies?’, For each reason, list only the respondent number.”, followed by respondent number and free-text answers in two columns, individually. The returned summarization and labeling of each answer were then manually inspected and adjusted (merging of reason 3 and 6). For question 50.1-5 (“What do you now know about ILC that you wish you could have heard from and discussed with your…”), all answers were combined ( N = 1260), with each answer transformed into a 384-dimentional vector (embedding) via sentence transformers ( https://huggingface.co/sentence-transformers ), and converted to 5-dimension using UMAP with local neighborhood at 15. HDBSCAN was then performed with minimum cluster size of 15, on Euclidean distance and cluster selection method as ‘eom’, generating 23 clusters. Then for each cluster, class-based TF-IDF (c-TF-IDF) vectors were calculated among all words, followed by topic reduction via merging c-TF-IDF vectors with highest cosine similarity with 10 iterations, generating 14 final topics. The resulted topics were merged into 11 via manual assessment of words of top frequency within each cluster, after which all answers of the same topic, except for ‘ambiguous’ group, were passed to gpt-4 api deterministic model (temperature zero) respectively, with a prefix “Think as a patient with breast cancer, and as a physician scientist studying breast cancer, summarize in professional scientific language one single common topic shared among answers, ‘What do you now know about ILC that you wish you could have heard from and discussed with your breast cancer doctor?’ The summarization should be a single, brief sentence”. Then metastasis and recurrence groups were manually merged based on expert suggestion. This eventually led to 10 topic clusters, including one non-specific group (ambiguous).
Crosstabulations were analyzed using Chi-Square test, and for these some groups were combined (such as pathologists, radiation oncologists, gynecologists, and others) due to the limited number in such subgroups analysis.
The study does not contain any sequencing or structural data. Majority of responses to survey as well as original survey document have been uploaded into Supplementary Text and Data Files. Additional raw data are available upon reasonable request from the corresponding author.
Codes for text analysis are provided in https://github.com/leeoesterreich/ILC-Survey .
National Program of Cancer Registries and Surveillance, Epidemiology, and End Results SEER*Stat Database: NPCR and SEER Incidence - U.S. Cancer Statistics 2001-2017 Public Use Research Database, 2019 Submission (2001-2017), United States Department of Health and Human Services, Centers for Disease Control and Prevention and National Cancer Institute. Released June 2020. Accessed at www.cdc.gov/cancer/uscs/public-use .
McCart Reed, A. E., Kalinowski, L., Simpson, P. T. & Lakhani, S. R. Invasive lobular carcinoma of the breast: the increasing importance of this special subtype. Breast Cancer Res. 23 , 6 (2021).
Article CAS PubMed PubMed Central Google Scholar
Christgen, M. & Derksen, P. Lobular breast cancer: molecular basis, mouse and cellular models. Breast Cancer Res. 17 , 16 (2015).
Article PubMed PubMed Central Google Scholar
De Schepper, M. et al. Results of a worldwide survey on the currently used histopathological diagnostic criteria for invasive lobular breast cancer. Mod. Pathol. 35 , 1812–1820 (2022).
Christgen, M. et al. Inter-observer agreement for the histological diagnosis of invasive lobular breast carcinoma. J. Pathol. Clin. Res. 8 , 191–205 (2022).
Article CAS PubMed Google Scholar
Ciriello, G. et al. Comprehensive Molecular Portraits of Invasive Lobular Breast Cancer. Cell 163 , 506–519 (2015).
Van Baelen, K. et al. Current and future diagnostic and treatment strategies for patients with invasive lobular breast cancer. Ann. Oncol. 33 , 769–785 (2022).
Oesterreich, S. et al. Clinicopathological features and outcomes comparing patients with invasive ductal and lobular breast cancer. J. Natl Cancer Inst. 114 , 1511–1522 (2022).
Marmor, S. et al. Relative effectiveness of adjuvant chemotherapy for invasive lobular compared with invasive ductal carcinoma of the breast. Cancer 123 , 3015–3021 (2017).
Truin, W. et al. Effect of adjuvant chemotherapy in postmenopausal patients with invasive ductal versus lobular breast cancer. Ann. Oncol. 23 , 2859–2865 (2012).
Hutcheson, L. B. et al. Abstract P3-15-03: Imaging and invasive lobular carcinoma: A survey study conducted by the lobular breast cancer alliance. Cancer Res. 82 , P3-15-03–P3-15-03 (2022).
Article Google Scholar
Pereslucha, A. M., Wenger, D. M., Morris, M. F. & Aydi, Z. B. Invasive lobular carcinoma: A review of imaging modalities with special focus on pathology concordance. Healthcare 11 , 746 (2023).
Gilardi, L. et al. FDG and Non-FDG Radiopharmaceuticals for PET imaging in invasive lobular breast carcinoma. Biomedicines 11 , 1350 (2023).
Ulaner, G. A. et al. Head-to-head evaluation of (18)F-FES and (18)F-FDG PET/CT in metastatic invasive lobular breast cancer. J. Nucl. Med. 62 , 326–331 (2021).
Sandbank, J. et al. Validation and real-world clinical application of an artificial intelligence algorithm for breast cancer detection in biopsies. NPJ Breast Cancer 8 , 129 (2022).
Reis-Filho, J. S. et al. Abstract PD11-01: An artificial intelligence-based predictor of CDH1 biallelic mutations and invasive lobular carcinoma. Cancer Res. 82 , PD11-01–PD11-01 (2022).
Christgen, M. et al. Lobular breast cancer: histomorphology and different concepts of a special spectrum of tumors. Cancers 13 , 3695 (2021).
Metzger Filho, O. et al. Relative eectiveness of Letrozole compared With Tamoxifen for patients with lobular carcinoma in the BIG 1-98 Trial. J. Clin. Oncol. 33 , 2772–2779 (2015).
Nardone, A. et al. A distinct chromatin state drives therapeutic resistance in invasive lobular breast cancer. Cancer Res. 82 , 3673–3686 (2022).
Sikora, M. J. et al. Invasive lobular carcinoma cell lines are characterized by unique estrogen-mediated gene expression patterns and altered tamoxifen response. Cancer Res 74 , 1463–1474 (2014).
Bajrami, I. et al. E-Cadherin/ROS1 inhibitor synthetic lethality in breast cancer. Cancer Discov. 8 , 498–515 (2018).
Elangovan, A. et al. Loss of E-cadherin Induces IGF1R activation and reveals a targetable pathway in invasive lobular breast carcinoma. Mol. Cancer Res. 20 , 1405–1419 (2022).
Nagle, A. M. et al. Loss of E-cadherin enhances IGF1-IGF1R pathway activation and sensitizes breast cancers to anti-IGF1R/InsR inhibitors. Clin. Cancer Res. 24 , 5165–5177 (2018).
Teo, K. et al. E-cadherin loss induces targetable autocrine activation of growth factor signalling in lobular breast cancer. Sci. Rep. 8 , 15454 (2018).
Hornsveld, M. et al. Restraining FOXO3-dependent transcriptional BMF activation underpins tumour growth and metastasis of E-cadherin-negative breast cancer. Cell Death Differ. 23 , 1483–1492 (2016).
Voorwerk, L. et al. PD-L1 blockade in combination with carboplatin as immune induction in metastatic lobular breast cancer: the GELATO trial. Nat. Cancer 4 , 535–549 (2023).
Onkar, S. et al. Immune landscape in invasive ductal and lobular breast cancer reveals a divergent macrophage-driven microenvironment. Nat. Cancer 4 , 516–534 (2023).
Download references
We thank all respondents to the survey who made this work possible. This work is dedicated to Leigh Pate, who was not only instrumental in the development of this survey, but who laid the foundation for worldwide lobular breast cancer advocacy. We would also like to thank many other patient advocates representing the different ILC advocacy groups such as LBCA and ELBCC patient advocates (ELBCA) who have contributed to editing the original survey questions. This work was made in part possible by the Survey core of the UPMC Hillman Cancer Center with support from NIH grant award P30CA047904. We would like to thank the Shear Family Foundation for their support of generation of the survey and analysis of results. Fangyuan Chen was a former visiting research scholar at the University of Pittsburgh School of Medicine supported by funds from The China Scholarship Council and Tsinghua University. The study is in part based on work from COST Action LOBSTERPOT (CA19138), supported by COST (European Cooperation in Science and Technology). COST (European Cooperation in Science and Technology) is a funding agency for research and innovation networks https://www.cost.eu . Research reported in this publication was supported in part by a Cancer Center Support Grant of the NIH/NCI (Grant No. P30CA008748; MSK). J.S. Reis-Filho was funded in part by Susan G Komen Leadership, and NIH/NCI P50 CA247749 01 grants. A number of authors are funded by the Breast Cancer Research Foundation (SO, AVL, NMD, CL, CS, CD, JSRF).
Jorge S. Reis-Filho
Present address: AstraZeneca, GAITHERSBURG, MARYLAND, USA
Deceased: Leigh Pate.
University of Pittsburgh, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Women’s Cancer Research Center, Magee Womens Research Institute, Pittsburgh, PA, USA
Steffi Oesterreich, Adrian V. Lee, Fangyuan Chen & Osama S. Shah
Independent ILC Advocate, Founder LBCA, Pittsburgh, PA, USA
Institute for Precision Medicine, University of Pittsburgh and UPMC, Pittsburgh, PA, USA
Adrian V. Lee
Tsinghua University, Beijing, China
Fangyuan Chen
University of Pennsylvania and Penn Medicine Abramson Cancer Center, Philadelphia, PA, USA
Rachel C. Jankowitz
University of California, San Francisco, Department of Surgery, San Francisco, CA, USA
Rita Mukhtar
Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA
Otto Metzger
Dept. of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
Matthew J. Sikora
Fred Hutchinson Cancer Center, Division of Public Health Sciences, Seattle, WA, USA
Christopher I. Li & Nancy M. Davidson
Jules Bordet Institute Belgium, Brussels, Belgium
Christos Sotiriou
Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
Thijs Koorman & Patrick Derksen
Molecular Imaging and Therapy, Hoag Family Cancer Institute, Molecular Imaging and Therapy, Irvine, CA. Departments of Radiology and Translational Genomics, University of Southern California, Los Angeles, CA, USA
Gary Ulaner
Memorial Sloan Kettering Cancer Center, New York, NY, USA
Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
Karen Van Baelen & Christine Desmedt
Lobular Breast Cancer Alliance Inc., White Horse Beach, MA, USA
Laurie Hutcheson
Lobular Ireland, Dublin, Ireland
Siobhan Freeney
Dynami Foundation, Wausau, WI, USA
Flora Migyanka
Lobular Breast Cancer UK, Nottingham, UK
Claire Turner
Department of Family Medicine, University of Pittsburgh, Pittsburgh, patients/advocates, Pittsburgh, PA, USA
You can also search for this author in PubMed Google Scholar
L.P., S.O. and A.V.L. conceptualized the study, and developed draft survey. Critical input on survey development including questions from R.C., R.M., O.M., M.J.S., C.L., C.S., T.K., G.U., J.S.R.F., N.M.D., K.V.B., L.H., S.F., F.M., C.T., P.D., T.B., C.D. Further refinement of survey by K.V.B. and C.D., together with S.O., A.V.L. and L.P. Setting up of online survey, and subsequent collection and analysis of survey data by T.B. Analysis of data also by F.C. and O.S.S. S.O., A.V.L., C.D. and L.P interpreted the data, and wrote the manscuript, and other major contributors in writing and figure generation were F.C., O.S.S. and T.D. All authors read and approved the final manuscript except L.P. who passed before finishing final draft of the manuscript.
Correspondence to Steffi Oesterreich .
Competing interests.
J.S. Reis-Filho reported receiving personal/consultancy fees from Goldman Sachs, Bain Capital, REPARE Therapeutics, Saga Diagnostics and Paige.AI, membership of the scientific advisory boards of VolitionRx, REPARE Therapeutics and Paige.AI, membership of the Board of Directors of Grupo Oncoclinicas, and ad hoc membership of the scientific advisory boards of AstraZeneca, Merck, Daiichi Sankyo, Roche Tissue Diagnostics and Personalis, outside the submitted work. The other authors indicated no potential conflicts of interest.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Suppl material, rights and permissions.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ .
Reprints and permissions
Cite this article.
Oesterreich, S., Pate, L., Lee, A.V. et al. International survey on invasive lobular breast cancer identifies priority research questions. npj Breast Cancer 10 , 61 (2024). https://doi.org/10.1038/s41523-024-00661-3
Download citation
Received : 23 January 2024
Accepted : 14 June 2024
Published : 20 July 2024
DOI : https://doi.org/10.1038/s41523-024-00661-3
Anyone you share the following link with will be able to read this content:
Sorry, a shareable link is not currently available for this article.
Provided by the Springer Nature SharedIt content-sharing initiative
Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.
Edited by Cancer Communications
Cancer Communications volume 38 , Article number: 69 ( 2018 ) Cite this article
23k Accesses
8 Citations
1 Altmetric
Metrics details
Since the beginning of 2017, Cancer Communications (former title: Chinese Journal of Cancer ) has published a series of important questions regarding cancer research and clinical oncology, to provide an enhanced stimulus for cancer research, and to accelerate collaborations between institutions and investigators. In this edition, the following 8 valuable questions are presented. Question 94. The origin of tumors: time for a new paradigm? Question 95. How can we accelerate the identification of biomarkers for the early detection of pancreatic ductal adenocarcinoma? Question 96. Can we improve the treatment outcomes of metastatic pancreatic ductal adenocarcinoma through precision medicine guided by a combination of the genetic and proteomic information of the tumor? Question 97. What are the parameters that determine a competent immune system that gives a complete response to cancers after immune induction? Question 98. Is high local concentration of metformin essential for its anti-cancer activity? Question 99. How can we monitor the emergence of cancer cells anywhere in the body through plasma testing? Question 100. Can phytochemicals be more specific and efficient at targeting P-glycoproteins to overcome multi-drug resistance in cancer cells? Question 101. Is cell migration a selectable trait in the natural evolution of carcinoma?
Until now, the battle against cancer is still ongoing, but there are also ongoing discoveries being made. Milestones in cancer research and treatments are being achieved every year; at a quicker pace, as compared to decades ago. Likewise, some cancers that were considered incurable are now partly curable, lives that could not be saved are now being saved, and for those with yet little options, they are now having best-supporting care. With an objective to promote worldwide cancer research and even accelerate inter-countries collaborations, since the beginning of 2017, Cancer Communications (former title: Chinese Journal of Cancer ) has launched a program of publishing 150 most important questions in cancer research and clinical oncology [ 1 ]. We are providing a platform for researchers to freely voice-out their novel ideas, and propositions to enhance the communications on how and where our focus should be placed [ 2 , 3 , 4 , 5 , 6 , 7 , 8 , 9 , 10 , 11 , 12 , 13 ]. In this edition, 8 valuable and inspiring questions, Question 94–101, from highly distinguished professionals from different parts of the world are presented. If you have any novel proposition(s) and Question(s), please feel free to contact Ms. Ji Ruan via email: [email protected].
Background and implications.
“There is no worse blind man than the one who doesn’t want to see. There is no worse deaf man than the one who doesn’t want to hear. And there is no worse madman than the one who doesn’t want to understand.” —Ancient Proverb
In the past half-century, cancer biologists have focused on a dogma in which cancer was viewed as a proliferative disease due to mechanisms that activate genes (oncogenes) to promote cell proliferation or inactivate genes (tumor suppressor genes) to suppress tumor growth. In retrospect, these concepts were established based on functional selections, by using tissue culture (largely mouse NIH 3T3 cells) for the selection of transformed foci at the time when we knew virtually nothing about the human genome [ 14 ]. However, it is very difficult to use these genes individually or in combinations to transform primary human cells. Further, the simplified view of uncontrolled proliferation cannot explain the tumor as being a malignant organ or a teratoma, as observed by pathologists over centuries. Recently, the cancer genomic atlas project has revealed a wide variety of genetic alterations ranging from no mutation to multiple chromosomal deletions or fragmentations, which make the identification of cancer driver mutations very challenging in a background of such a massive genomic rearrangement. Paradoxically, this increase the evidences demonstrating that the oncogenic mutations are commonly found in many normal tissues, further challenging the dogma that genetic alteration is the primary driver of this disease.
Logically, the birth of a tumor should undergo an embryonic-like development at the beginning, similar to that of a human. However, the nature of such somatic-derived early embryo has been elusive. Recently, we provided evidence to show that polyploid giant cancer cells (PGCCs), which have been previously considered non-dividing, are actually capable of self-renewal, generating viable daughter cells via amitotic budding, splitting and burst, and capable of acquisition of embryonic-like stemness [ 15 , 16 , 17 ]. The mode of PGCC division is remarkably similar to that of blastomere, a first step in human embryogenesis following fertilization. The blastomere nucleus continuously divides 4–5 times without cytoplasmic division to generate 16–32 cells and then to form compaction/morulae before developing into a blastocyst [ 18 ]. Based on these data and similarity to the earliest stage of human embryogenesis, I propose a new theory that tumor initiation can be achieved via a dualistic origin, similar to the first step of human embryogenesis via the formation of blastomere-like cells, i.e. the activation of blastomere or blastomere-like cells which leads to the dedifferentiation of germ cells or somatic cells, respectively, which is then followed by the differentiation to generate their respective stem cells, and the differentiation arrest at a specific developmental hierarchy leading to tumor initiation [ 19 ]. The somatic-derived blastomere-like cancer stem cell follows its own mode of cell growth and division and is named as the giant cell cycle. This cycle includes four distinct but overlapping phases: the initiation, self-renewal, termination, and stability phases. The giant cell cycle can be tracked in vitro and in vivo due to their salient giant cell morphology (Fig. 1 ).
One mononucleated polyploid giant cancer cell (PGCC) in the background of regular size diploid cancer cells. The PGCC can be seen to be at least 100 times larger than that of regular cancer cells
This new theory challenges the traditional paradigm that cancer is a proliferative disease, and proposes that the initiation of cancer requires blastomere-like division that is similar to that of humans before achieving stable proliferation at specific developmental hierarchy in at least half of all human cancers. This question calls for all investigators in the cancer research community to investigate the role of PGCCs in the initiation, progression, resistance, and metastasis of cancer and to look for novel agents to block the different stages of the giant cell cycle.
The histopathology (phenotype) of cancers has been there all the time. It is just the theory of cancer origin proposed by scientists that changes from time to time. After all, trillions of dollars have been invested in fighting this disease by basing on its genetic origin in the past half-century, yet, little insight has been gained [ 14 ]. Here are two quotes from Einstein: “Insanity: doing the same thing over and over again expecting different results”, and “We cannot solve our problems with the same thinking we used when created them”.
In short, it is time to change our mindset and to start pursuing PGCCs, which we can observe under the microscope. But with very little understanding about these cells, it is time for a shift in paradigm.
Jinsong Liu.
Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4095, USA.
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers in the world with a dismal 5-year overall survival rate of less than 5%; which has not been significantly improved since the past decades. Although surgical resection is the only option for curative treatment of PDAC, only 15%–20% of patients with PDAC have the chance to undergo curative resection, leaving the rest with only palliative options in hope for increasing their quality of life; since they were already at unresectable and non-curative stages at their first diagnosis.
The lack of specific symptoms in the early-stage of PDAC is responsible for rendering an early diagnosis difficult. Therefore, more sensitive and specific screening methodologies for its early detection is urgently needed to improve its diagnosis, starting early treatments, and ameliorating prognoses. The diagnosis so far relies on imaging modalities such as abdominal ultrasound, computed tomography (CT), magnetic resonance imaging (MRI), endoscopic ultrasound (EUS), endoscopic retrograde cholangiopancreatography (ERCP), and positron emission tomography (PET). One may propose to screen for pancreatic cancer in high-risk populations, which is highly recommended, however screening intervention for all the people is not a wise choice; when considering the relatively low prevalence of PDAC, and the difficulty for diagnosing it in its early stage [ 20 ].
Therefore, alternative diagnostic tools for early detection of PDAC are highly expected. Among the biomarkers currently used in clinical practice, carbohydrate antigen 19–9 (CA19–9) is among the most useful one for supporting the diagnosis of PDAC, but it is neither sufficiently sensitive nor specific for its early detection. Yachida et al. reported in 2010 that the initiating mutation in the pancreas occurs approximately two decades before the PDAC to start growing in distant organs [ 21 ], which indicates a broad time of the window of opportunity for the early detection of PDAC. With the advancement in next-generation sequencing technology, the number of reported studies regarding novel potential molecular biomarkers in bodily fluids including the blood, feces, urine, saliva, and pancreatic juice for early detection of PDAC has been increasing. Such biomarkers may be susceptible to detect mutations at the genetic or epigenetic level, identifying important non-coding RNA (especially microRNA and long non-coding RNA), providing insights regarding the metabolic profiles, estimating the tumor level in liquid biopsies (circulating free DNA, circulating tumor cells and exosomes), and so on.
Another approach to identifying biomarkers for the early detection of pancreatic cancer is using animal models. In spontaneous animal models of pancreatic cancer, such as Kras-mutated mouse models, it is expected that by high throughput analyses of the genetic/epigenetic/proteomic alterations, some novel biomarkers might be able to be identified. For instance, Sharma et al. reported in 2017 that the detection of phosphatidylserine-positive exosomes enabled the diagnosis of early-stage malignancies in LSL-Kras G12D , Cdkn2a lox/lox : p48 Cre and LSL-Kras G12d/+ , LSL-Trp R172H/+ , and P48 Cre mice [ 22 ].
These analyses in clinical samples or animal models hold the clues for the early detection of PDAC, however, further studies are required to validate their diagnostic performance. What’s most important, will be the lining-up of these identified prospective biomarkers, to validate their sensitivities and specificities. This will determine their potential for widespread clinical applicability, and hopefully, accelerate the early diagnosis of PDAC.
Mikiya Takao 1,2 , Hirotaka Matsuo 2 , Junji Yamamoto 1 , and Nariyoshi Shinomiya 2 .
1 Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan; 2 Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
[email protected]; [email protected]; [email protected]; [email protected]
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant cancers, and nearly half of the patients had metastatic PDAC when they are initially diagnosed. When they are accompanied by metastatic tumors, unlike most solid cancer, PDAC cannot be cured with primary surgical resection alone [ 23 , 24 ]. Also, since PDAC has poor responses to conventional therapies, improvements in adjunctive treatment approach including chemo- and immuno-therapy are earnestly required. From this standpoint, recent results regarding the differences in the molecular evolution of pancreatic cancer subtypes provide a new insight into its therapeutic development [ 25 ], which may lead to the improvement of the prognosis of not only metastatic PDAC but also of locally advanced or recurrent PDAC.
In fact, new chemotherapeutic regimens such as the combination of gemcitabine with nab-paclitaxel and FOLFIRINOX have been reported to show improved prognosis despite a lack of examples of past successes in the treatment of patients with metastatic PDAC who had undergone R0 resection [ 26 ]. While many mutations including KRAS , CDKN2A , TP53, and SMAD4 are associated with pancreatic carcinogenesis, no effective molecular targeted drug has been introduced in the clinical setting so far. A recent report of a phase I/II study on refametinib, a MEK inhibitor, indicated that KRAS mutation status might affect the overall response rate, disease control rate, progression-free survival, and overall survival of PDAC in combination with gemcitabine [ 27 ].
While immunotherapy is expected to bring a great improvement in cancer treatment, until now, immune checkpoint inhibitors have achieved limited clinical benefit for patients with PDAC. This might be because PDAC creates a uniquely immunosuppressive tumor microenvironment, where tumor-associated immunosuppressive cells and accompanying desmoplastic stroma prevent the tumor cells from T cell infiltration. Recently reported studies have indicated that immunotherapy might be effective when combined with focal adhesion kinase (FAK) inhibitor [ 28 ] or IL-6 inhibitor [ 29 ], but more studies are required to validate their use in clinical practice.
As such, we believe that if the dynamic monitoring of drug sensitivity/resistance in the individual patients is coupled with precision treatment based on individualized genetics/epigenetics/proteomics alterations in the patients’ tumor, this could improve the treatment outcomes of PDAC.
Mikiya Takao 1,2 , Hirotaka Matsuo 2 , Junji Yamamoto 1 , and Nariyoshi Shinomiya 2.
Recently, cancer immunotherapy has shown great clinical benefit in multiple types of cancers [ 30 , 31 , 32 ]. It has provided new approaches for cancer treatment. However, it has been observed that only a fraction of patients respond to immunotherapy.
Much effort has been made to identify markers for immunotherapeutic response. Tumor mutation burden (TMB), mismatch repair (MMR) deficiency, PD-L1 expression, and tumor infiltration lymphocyte (TIL) have been found to be associated with an increased response rate in checkpoint blockade therapies. Unfortunately, a precise prediction is still challenging in this field. Moreover, when to stop the treatment of immunotherapy is an urgent question that remains to be elucidated.
In other words, there is no available approach to determine if a patient has generated a good immune response against the cancer after immunotherapy treatments. All of these indicate the complexity and challenges that reside for implementing novel man-induced cancer-effective immune response therapeutics. A variety of immune cells play collaborative roles at different stages to recognize antigens and eventually to generate an effective anti-cancer immune response. Given the high complexity of the immune system, a rational evaluation approach is needed to cover the whole process. Moreover, we need to perfect vaccine immunization and/or in vitro activation of T cells to augment the function of the immune system; particularly the formation of immune memory.
Edison Liu 1 , Penghui Zhou 2 , Jiang Li 2 .
1 The Jackson Laboratory, Bar Harbor, ME 04609, USA; 2 Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P. R. China.
[email protected]; [email protected]; [email protected]
Metformin was approved as a first line of anti-diabetic drug since decades. Interestingly, the fact that clinical epidemiological studies have shown that metformin can reduce the risk of a variety of cancers stimulates considerable recognition to explore its anticancer activity.
Although the in vitro and in vivo experimental results have demonstrated that metformin can have some potential anti-tumor effects, more than 100 clinical trials did not achieve such desirable results [ 33 ]. We and others believe that the main problem resides in the prescribing doses used. For cancer treatment, a much higher dose may be needed for observing any anti-tumor activities, as compared to the doses prescribed for diabetics [ 34 , 35 , 36 ].
Further, if the traditional local/oral administration approach is favored, the prescribed metformin may not be at the required dose-concentration once it reaches the blood to have the effective anti-cancer activities. We, therefore, propose that intravesical instillation of metformin into the bladder lumen could be a promising way to treat for bladder cancer, at least. We have already obtained encouraging results both in vitro and in vivo experiments, including in an orthotopical bladder cancer model [ 36 , 37 ]. Now, we are waiting to observe its prospective clinical outcome.
Mei Peng 1 , Xiaoping Yang 2 .
1 Department of Pharmacy, Xiangya Hospital, Central South University. Changsha, Hunan 410083, P. R. China; 2 Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P. R. China.
[email protected]; [email protected]
The early detection of cancer is still a relentless worldwide challenge. The sensitivity and specificity of traditional blood tumor markers and imaging technologies are still to be greatly improved. Hence, novel approaches for the early detection of cancer are urgently needed.
The emergence of liquid biopsy technologies opens a new driveway for solving such issues. According to the definition of the National Cancer Institute of the United States, a liquid biopsy is a test done on a sample of blood to look for tumorigenic cancer cells or pieces of tumor cells’ DNA that are circulating in the blood [ 38 ]. This definition implies two main types of the current liquid biopsy: one that detects circulating tumor cells and the other that detects non-cellular material in the blood, including tumor DNA, RNA, and exosomes.
Circulating tumor cells (CTCs) are referred to as tumor cells that have been shed from the primary tumor location and have found their way to the peripheral blood. CTCs were first described in 1869 by an Australian pathologist, Thomas Ashworth, in a patient with metastatic cancer [ 39 ]. The importance of CTCs in modern cancer research began in the mid-1990s with the demonstration that CTCs exist early in the course of the disease.
It is estimated that there are about 1–10 CTCs per mL in whole blood of patients with metastatic cancer, even fewer in patients with early-stage cancer [ 40 ]. For comparison, 1 mL of blood contains a few million white blood cells and a billion erythrocytes. The identification of CTCs, being in such low frequency, requires some special tumoral markers (e.g., EpCAM and cytokeratins) to capture and isolate them. Unfortunately, the common markers for recognizing the majority of CTCs are not effective enough for clinical application [ 41 ]. Although accumulated evidences have shown that the presence of CTCs is a strong negative prognostic factor in the patients with metastatic breast, lung and colorectal cancers, detecting CTCs might not be an ideal branch to hold on for the hope of early cancer detection [ 42 , 43 , 44 , 45 ].
Circulating tumor DNA (ctDNA) is tumor-derived fragmented DNA in the circulatory system, which is mainly derived from the tumor cell death through necrosis and/or apoptosis [ 46 ]. Given its origin, ctDNA inherently carries cancer-specific genetic and epigenetic aberrations, which can be used as a surrogate source of tumor DNA for cancer diagnosis and prognostic prediction. Ideally, as a noninvasive tumor early screening tool, a liquid biopsy test should be able to detect many types of cancers and provide the information of tumor origin for further specific clinical management. In fact, the somatic mutations of ctDNA in different types of tumor are highly variable, even in the different individuals with the same type of tumor [ 47 ]. Additionally, most tumors do not possess driver mutations, with some notable exceptions, which make the somatic mutations of ctDNA not suitable for early detection of the tumor.
Increased methylation of the promoter regions of tumor suppressor genes is an early event in many types of tumor, suggesting that altered ctDNA methylation patterns could be one of the first detectable neoplastic changes associated with tumorigenesis [ 48 ]. ctDNA methylation profiling provides several advantages over somatic mutation analysis for cancer detection including higher clinical sensitivity and dynamic range, multiple detectable methylation target regions, and multiple altered CpG sites within each targeted genomic region. Further, each methylation marker is present in both cancer tissue and ctDNA, whereas only a fraction of mutations present in cancer tissue could be detected in ctDNA.
In 2017, there were two inspiring studies that revealed the values of using ctDNA methylation analysis for cancer early diagnosis [ 49 , 50 ]. After partitioning the human genome into blocks of tightly coupled CpG methylation sites, namely methylation haplotype blocks (MHBs), Guo and colleagues performed tissue-specific methylation analyses at the MHBs level to accurately determine the tissue origin of the cancer using ctDNA from their enrolled patients [ 49 ]. In another study, Xu and colleagues identified a hepatocellular carcinoma (HCC) enriched methylation marker panel by comparing the HCC tissue and blood leukocytes from normal individuals and showed that methylation profiles of HCC tumor DNA and matched plasma ctDNA were highly correlated. In this study, after quantitative measurement of the methylation level of candidate markers in ctDNA from a large cohort of 1098 HCC patients and 835 normal controls, ten methylation markers were selected to construct a diagnostic prediction model. The proposed model demonstrated a high diagnostic specificity and sensitivity, and was highly correlated with tumor burden, treatment response, and tumor stage [ 50 ].
With the rapid development of highly sensitive detection methods, especially the technologies of massively parallel sequencing or next-generation sequencing (NGS)-based assays and digital PCR (dPCR), we strongly believe that the identification of a broader “pan-cancer” methylation panel applied for ctDNA analyses, probably in combination with detections of somatic mutation and tumor-derived exosomes, would allow more effective screening for common cancers in the near future.
Edison Liu 1 , Hui-Yan Luo 2 .
[email protected]; [email protected]
Though several anticancer agents are approved to treat different types of cancers, their full potentials have been limited due to the occurrence of drug resistance. Resistance to anticancer drugs develops by a variety of mechanisms, one of which is increased drug efflux by transporters. The ATP-binding cassette (ABC) family drug efflux transporter P-glycoprotein (P-gp or multi-drug resistance protein 1 [MDRP1]) has been extensively studied and is known to play a major role in the development of multi-drug resistance (MDR) to chemotherapy [ 51 ]. In brief, overexpressed P-gp efflux out a wide variety of anticancer agents (e.g.: vinca alkaloids, doxorubicin, paclitaxel, etc.), leading to a lower concentration of these drugs inside cancer cells, thereby resulting in MDR. Over the past three decades, researchers have developed several synthetic P-gp inhibitors to block the efflux of anticancer drugs and have tested them in clinical trials, in combination with chemotherapeutic drugs. But none were found to be suitable enough in overcoming MDR and to be released for marketing, mainly due to the side effects associated with cross-reactivity towards other ABC transporters (BCRP and MRP-1) and the inhibition of CYP450 drug metabolizing enzymes [ 52 , 53 ].
On the other hand, a number of phytochemicals have been reported to have P-gp inhibitory activity. Moreover, detailed structure–activity studies on these phytochemicals have delineated the functional groups essential for P-gp inhibition [ 53 , 54 ]. Currently, one of the phytochemicals, tetrandrine (CBT-1 ® ; NSC-77037), is being used in a Phase I clinical trial ( http://www.ClinicalTrials.gov ; NCT03002805) in combination with doxorubicin for the treatment of metastatic sarcoma. Before developing phytochemicals or their derivatives as P-gp inhibitors, they need to be investigated thoroughly for their cross-reactivity towards other ABC transporters and CYP450 inhibition, in order to avoid toxicities similar to the older generation P-gp inhibitors that have failed in clinical trials.
Therefore, the selectivity for P-gp over other drug transporters and drug metabolizing enzymes should be considered as important criterias for the development of phytochemicals and their derivatives for overcoming MDR.
Mohane Selvaraj Coumar and Safiulla Basha Syed.
Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India.
[email protected]; [email protected]
The propensity of solid tumor malignancy to metastasize remains the main cause of cancer-related death, an extraordinary unmet clinical need, and an unanswered question in basic cancer research. While dissemination has been traditionally viewed as a late process in the progression of malignant tumors, amount of evidence indicates that it can occur early in the natural history of cancer, frequently when the primary lesion is still barely detectable.
A prerequisite for cancer dissemination is the acquisition of migratory/invasive properties. However, whether, and if so, how the migratory phenotype is selected for during the natural evolution of cancer and what advantage, if any, it may provide to the growing malignant cells remains an open issue. The answers to these questions are relevant not only for our understating of cancer biology but also for the strategies we adopt in an attempt of curbing this disease. Frequently, indeed, particularly in pharmaceutical settings, targeting migration has been considered much like trying “to shut the stable door after the horse has bolted” and no serious efforts in pursuing this aim has been done.
We argue, instead, that migration might be an intrinsic cancer trait that much like proliferation or increased survival confers to the growing tumor masses with striking selective advantages. The most compelling evidence in support for this contention stems from studies using mathematical modeling of cancer evolution. Surprisingly, these works highlighted the notion that cell migration is an intrinsic, selectable property of malignant cells, so intimately intertwined with more obvious evolutionarily-driven cancer traits to directly impact not only on the potential of malignant cells to disseminate but also on their growth dynamics, and ultimately provide a selective evolutionary advantage. Whether in real life this holds true remains to be assessed, nevertheless, work of this kind defines a framework where the acquisition of migration can be understood in a term of not just as a way to spread, but also to trigger the emergence of malignant clones with favorable genetic or epigenetic traits.
Alternatively, migratory phenotypes might emerge as a response to unfavorable conditions, including the mechanically challenging environment which tumors, and particularly epithelial-derived carcinoma, invariably experience. Becoming motile, however, may not per se being fixed as phenotypic advantageous traits unless it is accompanied or is causing the emergence of specific traits, including drug resistance, self-renewal, and survival. This might be the case, for example, during the process of epithelial-to-mesenchymal transition (EMT), which is emerging as an overarching mechanism for dissemination. EMT, indeed, may transiently equip individual cancer cells not only with migratory/invasive capacity but also with increased resistance to drug treatment, stemness potential at the expanse of fast proliferation.
Thus, within this framework targeting pro-migratory genes, proteins and processes may become a therapeutically valid alternative or a complementary strategy not only to control carcinoma dissemination but also its progression and development.
Giorgio Scita.
IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), School of Medicine, University of Milan, Via Festa del Perdono 7, 20122, Italy.
Qian CN, Zhang W, Xu RH. Defeating cancer: the 150 most important questions in cancer research and clinical oncology. Chin J Cancer. 2016;35(1):104. https://doi.org/10.1186/s40880-016-0165-4 .
Article PubMed PubMed Central Google Scholar
Wee JT, Poh SS. The most important questions in cancer research and clinical oncology: question 1. Could the vertical transmission of human papilloma virus (HPV) infection account for the cause, characteristics, and epidemiology of HPV-positive oropharyngeal carcinoma, non-smoking East Asian female lung adenocarcinoma, and/or East Asian triple-negative breast carcinoma? Chin J Cancer. 2017;36(1):13. https://doi.org/10.1186/s40880-016-0168-1 .
Venniyoor A. The most important questions in cancer research and clinical oncology—Question 2–5. Obesity-related cancers: more questions than answers. Chin J Cancer. 2017;36(1):18. https://doi.org/10.1186/s40880-017-0185-8 .
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 6–14: Edited by Chinese Journal of Cancer. Chin J Cancer. 2017;36(1):33. https://doi.org/10.1186/s40880-017-0200-0 .
Article Google Scholar
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 15–24: Edited by Chinese Journal of Cancer. Chin J Cancer. 2017;36(1):39. https://doi.org/10.1186/s40880-017-0205-8 .
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 25–30: Edited by Chinese Journal of Cancer. Chin J Cancer. 2017;36(1):42. https://doi.org/10.1186/s40880-017-0210-y .
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 31–39: Edited by Chinese Journal of Cancer. Chin J Cancer. 2017;36(1):48. https://doi.org/10.1186/s40880-017-0215-6 .
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 40–49. Chin J Cancer. 2017;36(1):55. https://doi.org/10.1186/s40880-017-0222-7 .
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 50–56. Chin J Cancer. 2017;36(1):69. https://doi.org/10.1186/s40880-017-0236-1 .
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 57–66: Edited by Chinese Journal of Cancer. Chin J Cancer. 2017;36(1):79. https://doi.org/10.1186/s40880-017-0249-9 .
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 67–75: Edited by Chinese Journal of Cancer. Chin J Cancer. 2017;36(1):86. https://doi.org/10.1186/s40880-017-0254-z .
Editorial Office of Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 76–85: Edited by Chinese Journal of Cancer. Chin J Cancer. 2017;36(1):91. https://doi.org/10.1186/s40880-017-0259-7 .
Chinese Journal of C. The 150 most important questions in cancer research and clinical oncology series: questions 86–93: Edited by Chinese Journal of Cancer. Chin J Cancer. 2018;37(1):1. https://doi.org/10.1186/s40880-018-0266-3 .
Weinberg RA. Coming full circle-from endless complexity to simplicity and back again. Cell. 2014;157(1):267–71. https://doi.org/10.1016/j.cell.2014.03.004 .
Article CAS PubMed Google Scholar
Niu N, Mercado-Uribe I, Liu J. Dedifferentiation into blastomere-like cancer stem cells via formation of polyploid giant cancer cells. Oncogene. 2017;36(34):4887–900. https://doi.org/10.1038/onc.2017.72 .
Article CAS PubMed PubMed Central Google Scholar
Niu N, Zhang J, Zhang N, Mercado-Uribe I, Tao F, Han Z, et al. Linking genomic reorganization to tumor initiation via the giant cell cycle. Oncogenesis. 2016;5(12):e281. https://doi.org/10.1038/oncsis.2016.75 .
Zhang S, Mercado-Uribe I, Xing Z, Sun B, Kuang J, Liu J. Generation of cancer stem-like cells through the formation of polyploid giant cancer cells. Oncogene. 2014;33(1):116–28. https://doi.org/10.1038/onc.2013.96 .
Hemberger M, Dean W, Reik W. Epigenetic dynamics of stem cells and cell lineage commitment: digging Waddington’s canal. Nat Rev Mol Cell Biol. 2009;10(8):526–37. https://doi.org/10.1038/nrm2727 .
Liu J. The dualistic origin of human tumors. Semin Cancer Biol. 2018. https://doi.org/10.1016/j.semcancer.2018.07.004 .
Zhou B, Xu JW, Cheng YG, Gao JY, Hu SY, Wang L, et al. Early detection of pancreatic cancer: where are we now and where are we going? Int J Cancer. 2017;141(2):231–41. https://doi.org/10.1002/ijc.30670 .
Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467(7319):1114–7. https://doi.org/10.1038/nature09515 .
Sharma R, Huang X, Brekken RA, Schroit AJ. Detection of phosphatidylserine-positive exosomes for the diagnosis of early-stage malignancies. Br J Cancer. 2017;117(4):545–52. https://doi.org/10.1038/bjc.2017.183 .
Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med. 2014;371(11):1039–49. https://doi.org/10.1056/NEJMra1404198 .
Takada T, Yasuda H, Amano H, Yoshida M, Uchida T. Simultaneous hepatic resection with pancreato-duodenectomy for metastatic pancreatic head carcinoma: does it improve survival? Hepatogastroenterology. 1997;44(14):567–73.
CAS PubMed Google Scholar
Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531(7592):47–52. https://doi.org/10.1038/nature16965 .
Frigerio I, Regi P, Giardino A, Scopelliti F, Girelli R, Bassi C, et al. Downstaging in stage IV pancreatic cancer: a new population eligible for surgery? Ann Surg Oncol. 2017;24(8):2397–403. https://doi.org/10.1245/s10434-017-5885-4 .
Article PubMed Google Scholar
Van Laethem JL, Riess H, Jassem J, Haas M, Martens UM, Weekes C, et al. Phase I/II study of refametinib (BAY 86-9766) in combination with gemcitabine in advanced pancreatic cancer. Target Oncol. 2017;12(1):97–109. https://doi.org/10.1007/s11523-016-0469-y .
Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med. 2016;22(8):851–60. https://doi.org/10.1038/nm.4123 .
Mace TA, Shakya R, Pitarresi JR, Swanson B, McQuinn CW, Loftus S, et al. IL-6 and PD-L1 antibody blockade combination therapy reduces tumour progression in murine models of pancreatic cancer. Gut. 2018;67(2):320–32. https://doi.org/10.1136/gutjnl-2016-311585 .
Immunotherapy Beats Chemo for Bladder Cancer. Cancer Discov. 2017;7(5):OF8. https://doi.org/10.1158/2159-8290.cd-nb2017-035 .
Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018;19(5):603–15. https://doi.org/10.1016/S1470-2045(18)30142-6 .
Garassino MC, Cho BC, Kim JH, Mazieres J, Vansteenkiste J, Lena H, et al. Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): an open-label, single-arm, phase 2 study. Lancet Oncol. 2018;19(4):521–36. https://doi.org/10.1016/S1470-2045(18)30144-X .
U.S. National Library of Medicine. ClinicalTrials.gov.
Liu Z, Yokoyama NN, Blair CA, Li X, Avizonis D, Wu XR, et al. High sensitivity of an Ha-RAS transgenic model of superficial bladder cancer to metformin is associated with approximately 240-fold higher drug concentration in urine than serum. Mol Cancer Ther. 2016;15(3):430–8. https://doi.org/10.1158/1535-7163.MCT-15-0714-T .
Menendez JA, Quirantes-Pine R, Rodriguez-Gallego E, Cufi S, Corominas-Faja B, Cuyas E, et al. Oncobiguanides: paracelsus’ law and nonconventional routes for administering diabetobiguanides for cancer treatment. Oncotarget. 2014;5(9):2344–8. https://doi.org/10.18632/oncotarget.1965 .
Peng M, Su Q, Zeng Q, Li L, Liu Z, Xue L, et al. High efficacy of intravesical treatment of metformin on bladder cancer in preclinical model. Oncotarget. 2016;7(8):9102–17. https://doi.org/10.18632/oncotarget.6933 .
Peng M, Huang Y, Tao T, Peng CY, Su Q, Xu W, et al. Metformin and gefitinib cooperate to inhibit bladder cancer growth via both AMPK and EGFR pathways joining at Akt and Erk. Sci Rep. 2016;6:28611. https://doi.org/10.1038/srep28611 .
Definition of liquid biopsy. In: NCI Dictionary of Cancer Terms. National Cancer Institute. https://www.cancer.gov/publications/dictionaries/cancer-terms/def/liquid-biopsy .
Ta A. A case of cancer in which cells similar to those in the tumors were seen in the blood after death. Aust Med J. 1869;14:146–9.
Google Scholar
Miller MC, Doyle GV, Terstappen LW. Significance of circulating tumor cells detected by the cell search system in patients with metastatic breast colorectal and prostate cancer. J Oncol. 2010;2010:617421. https://doi.org/10.1155/2010/617421 .
Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450(7173):1235–9. https://doi.org/10.1038/nature06385 .
Bidard FC, Proudhon C, Pierga JY. Circulating tumor cells in breast cancer. Mol Oncol. 2016;10(3):418–30. https://doi.org/10.1016/j.molonc.2016.01.001 .
Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26(19):3213–21. https://doi.org/10.1200/JCO.2007.15.8923 .
Ignatiadis M, Dawson SJ. Circulating tumor cells and circulating tumor DNA for precision medicine: dream or reality? Ann Oncol. 2014;25(12):2304–13. https://doi.org/10.1093/annonc/mdu480 .
Krebs MG, Sloane R, Priest L, Lancashire L, Hou JM, Greystoke A, et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol. 2011;29(12):1556–63. https://doi.org/10.1200/JCO.2010.28.7045 .
Diaz LA Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32(6):579–86. https://doi.org/10.1200/JCO.2012.45.2011 .
Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502(7471):333–9. https://doi.org/10.1038/nature12634 .
Esteller M. Epigenetics in cancer. N Engl J Med. 2008;358(11):1148–59. https://doi.org/10.1056/NEJMra072067 .
Guo S, Diep D, Plongthongkum N, Fung HL, Zhang K, Zhang K. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat Genet. 2017;49(4):635–42. https://doi.org/10.1038/ng.3805 .
Xu RH, Wei W, Krawczyk M, Wang W, Luo H, Flagg K, et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater. 2017;16(11):1155–61. https://doi.org/10.1038/nmat4997 .
Robey RW, Pluchino KM, Hall MD, Fojo AT, Bates SE, Gottesman MM. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat Rev Cancer. 2018. https://doi.org/10.1038/s41568-018-0005-8 .
Chung FS, Santiago JS, Jesus MF, Trinidad CV, See MF. Disrupting P-glycoprotein function in clinical settings: what can we learn from the fundamental aspects of this transporter? Am J Cancer Res. 2016;6(8):1583–98.
CAS PubMed PubMed Central Google Scholar
Syed SB, Coumar MS. P-Glycoprotein mediated multidrug resistance reversal by phytochemicals: a review of SAR & future perspective for drug design. Curr Top Med Chem. 2016;16(22):2484–508.
Abdallah HM, Al-Abd AM, El-Dine RS, El-Halawany AM. P-Glycoprotein inhibitors of natural origin as potential tumor chemo-sensitizers: a review. J Adv Res. 2015;6(1):45–62. https://doi.org/10.1016/j.jare.2014.11.008 .
Download references
Authors and affiliations, rights and permissions.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/ ), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated.
Reprints and permissions
Cite this article.
Cancer Communications. The 150 most important questions in cancer research and clinical oncology series: questions 94–101. Cancer Commun 38 , 69 (2018). https://doi.org/10.1186/s40880-018-0341-9
Download citation
Received : 13 November 2018
Accepted : 19 November 2018
Published : 26 November 2018
DOI : https://doi.org/10.1186/s40880-018-0341-9
Anyone you share the following link with will be able to read this content:
Sorry, a shareable link is not currently available for this article.
Provided by the Springer Nature SharedIt content-sharing initiative
ISSN: 2523-3548
About Breast Cancer > What is Breast Cancer? > Breast Cancer Facts & Stats
Last updated on Aug 1, 2024
1 in 8 women in the United States will be diagnosed with breast cancer in her lifetime. In 2024, an estimated 310,720 women and 2,800 men will be diagnosed with invasive breast cancer. Chances are, you know at least one person who has been personally affected by breast cancer.
But there is hope. When caught in its earliest, localized stages, the 5-year relative survival rate is 99%. Advances in early detection and treatment methods have significantly increased breast cancer survival rates in recent years, and there are currently over 4 million breast cancer survivors in the United States.
Awareness of the facts and statistics surrounding breast cancer in the United States is key in empowering individuals to make informed decisions about their health.
Facts & statistics Incidence statistics Statistics by age Statistics by ethnicity Survival & mortality statistics Male breast cancer statistics Facts & statistics images
Breast cancer is a disease in which malignant (cancer) cells form in the tissues of the breast. There are many different types of breast cancer that can affect both women and men.
To determine the extent of an individual’s breast cancer and if it has spread outside of the breast, the cancer is assigned a stage upon diagnosis . The early detection of breast cancer through annual mammography and other breast exams is the best defense against receiving a late-stage breast cancer diagnosis. Generally speaking, the earlier the cancer is detected, the greater the likelihood of a successful outcome.
Though breast cancer in the United States occurs primarily in middle-aged and older women, age is not the only risk factor for a breast cancer diagnosis. Many risk factors beyond age may contribute to a breast cancer diagnosis, and sometimes there are no discernable risk factors at all.
In the United States, breast cancer occurs within every racial and ethnic group. However, there are variations in statistics and outcomes across the different groups. Learn more about how NBCF is addressing disparities in breast cancer .
Black Women:
Hispanic Women:
Asian, Pacific Islander, American Indian, and Alaska Native Women:
Breast cancer survival rates are calculated using different forms of data, including the type and staging of breast cancer at diagnosis. These rates give an idea of what percentage of people with the same type and stage of cancer are still alive after a certain time period—usually 5 years—after they were diagnosed. This is called the 5-year relative survival rate.
** (invasive cancer has not spread outside of the breast) | 99% |
(cancer has spread outside of the breast to nearby structures or lymph nodes) | 86% |
(cancer has spread to other parts of the body, such as lungs, liver, or bones) | 31% |
91% |
All people are born with some breast cells and tissue, including men. Although rare, men get breast cancer too .
Awareness is the first step in making informed choices about breast health. Donate now to help NBCF support more women and men facing breast cancer in communities throughout the United States.
Click on each image to download
Sources: 1 American Cancer Society ( cancer.org ) 2 Johns Hopkins ( hopkinsmedicine.org ) 3 National Cancer Institute ( cancer.gov ) 4 BreastCancer.org ( breastcancer.org ) 5 Centers for Disease Control & Prevention ( cdc.gov ) 6 Mayo Clinic ( mayoclinic.org ) 7 National Institutes of Health ( nih.gov )
Your Account
Manage your account, subscriptions and profile.
MyKomen Health
ShareForCures
In Your Community
In Your Community
View resources and events in your local community.
Change your location:
Susan G. Komen®
One moment can change everything.
Established and probable risk factors.
|
|
|
|
|
|
|
and inherited gene mutations and cancer risk |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Early and locally advanced breast cancer.
|
|
|
|
|
|
|
|
|
|
Metastatic breast cancer.
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Updated 10/04/24
Fact sheets
Breast cancer is a disease in which abnormal breast cells grow out of control and form tumours. If left unchecked, the tumours can spread throughout the body and become fatal.
Breast cancer cells begin inside the milk ducts and/or the milk-producing lobules of the breast. The earliest form (in situ) is not life-threatening and can be detected in early stages. Cancer cells can spread into nearby breast tissue (invasion). This creates tumours that cause lumps or thickening.
Invasive cancers can spread to nearby lymph nodes or other organs (metastasize). Metastasis can be life-threatening and fatal.
Treatment is based on the person, the type of cancer and its spread. Treatment combines surgery, radiation therapy and medications.
In 2022, there were 2.3 million women diagnosed with breast cancer and 670 000 deaths globally. Breast cancer occurs in every country of the world in women at any age after puberty but with increasing rates in later life. Global estimates reveal striking inequities in the breast cancer burden according to human development. For instance, in countries with a very high Human Development Index (HDI), 1 in 12 women will be diagnosed with breast cancer in their lifetime and 1 in 71 women die of it.
In contrast, in countries with a low HDI; while only 1 in 27 women is diagnosed with breast cancer in their lifetime, 1 in 48 women will die from it.
Female gender is the strongest breast cancer risk factor. Approximately 99% of breast cancers occur in women and 0.5–1% of breast cancers occur in men. The treatment of breast cancer in men follows the same principles of management as for women.
Certain factors increase the risk of breast cancer including increasing age, obesity, harmful use of alcohol, family history of breast cancer, history of radiation exposure, reproductive history (such as age that menstrual periods began and age at first pregnancy), tobacco use and postmenopausal hormone therapy. Approximately half of breast cancers develop in women who have no identifiable breast cancer risk factor other than gender (female) and age (over 40 years).
Family history of breast cancer increases the risk of breast cancer, but most women diagnosed with breast cancer do not have a known family history of the disease. Lack of a known family history does not necessarily mean that a woman is at reduced risk.
Certain inherited high penetrance gene mutations greatly increase breast cancer risk, the most dominant being mutations in the genes BRCA1, BRCA2 and PALB-2. Women found to have mutations in these major genes may consider risk reduction strategies such as surgical removal of both breasts or chemoprevention strategies.
Most people will not experience any symptoms when the cancer is still early hence the importance of early detection.
Breast cancer can have combinations of symptoms, especially when it is more advanced. Symptoms of breast cancer can include:
People with an abnormal breast lump should seek medical care, even if the lump does not hurt.
Most breast lumps are not cancer. Breast lumps that are cancerous are more likely to be successfully treated when they are small and have not spread to nearby lymph nodes.
Breast cancers may spread to other areas of the body and trigger other symptoms. Often, the most common first detectable site of spread is to the lymph nodes under the arm although it is possible to have cancer-bearing lymph nodes that cannot be felt.
Over time, cancerous cells may spread to other organs including the lungs, liver, brain and bones. Once they reach these sites, new cancer-related symptoms such as bone pain or headaches may appear.
Treatment for breast cancer depends on the subtype of cancer and how much it has spread outside of the breast to lymph nodes (stages II or III) or to other parts of the body (stage IV).
Doctors combine treatments to minimize the chances of the cancer coming back (recurrence). These include:
Treatments for breast cancer are more effective and are better tolerated when started early and taken to completion.
Surgery may remove just the cancerous tissue (called a lumpectomy) or the whole breast (mastectomy). Surgery may also remove lymph nodes to assess the cancer’s ability to spread.
Radiation therapy treats residual microscopic cancers left behind in the breast tissue and/or lymph nodes and minimizes the chances of cancer recurring on the chest wall.
Advanced cancers can erode through the skin to cause open sores (ulceration) but are not necessarily painful. Women with breast wounds that do not heal should seek medical care to have a biopsy performed.
Medicines to treat breast cancers are selected based on the biological properties of the cancer as determined by special tests (tumour marker determination). The great majority of drugs used for breast cancer are already on the WHO Essential Medicines List (EML).
Lymph nodes are removed at the time of cancer surgery for invasive cancers. Complete removal of the lymph node bed under the arm (complete axillary dissection) in the past was thought to be necessary to prevent the spread of cancer. A smaller lymph node procedure called “sentinel node biopsy” is now preferred as it has fewer complications.
Medical treatments for breast cancers, which may be given before (“neoadjuvant”) or after (“adjuvant”) surgery, is based on the biological subtyping of the cancers. Certain subtypes of breast cancer are more aggressive than others such as triple negative (those that do not express estrogen receptor (ER), progesterone receptor (PR) or HER-2 receptor). Cancer that express the estrogen receptor (ER) and/or progesterone receptor (PR) are likely to respond to endocrine (hormone) therapies such as tamoxifen or aromatase inhibitors. These medicines are taken orally for 5–10 years and reduce the chance of recurrence of these “hormone-positive” cancers by nearly half. Endocrine therapies can cause symptoms of menopause but are generally well tolerated.
Cancers that do not express ER or PR are “hormone receptor negative” and need to be treated with chemotherapy unless the cancer is very small. The chemotherapy regimens available today are very effective in reducing the chances of cancer spread or recurrence and are generally given as outpatient therapy. Chemotherapy for breast cancer generally does not require hospital admission in the absence of complications.
Breast cancers that independently overexpress a molecule called the HER-2/neu oncogene (HER-2 positive) are amenable to treatment with targeted biological agents such as trastuzumab. When targeted biological therapies are given, they are combined with chemotherapy to make them effective at killing cancer cells.
Radiotherapy plays a very important role in treating breast cancer. With early-stage breast cancers, radiation can prevent a woman having to undergo a mastectomy. With later stage cancers, radiotherapy can reduce cancer recurrence risk even when a mastectomy has been performed. For advanced stages of breast cancer, in some circumstances, radiation therapy may reduce the likelihood of dying of the disease.
The effectiveness of breast cancer therapies depends on the full course of treatment. Partial treatment is less likely to lead to a positive outcome.
Age-standardized breast cancer mortality in high-income countries dropped by 40% between the 1980s and 2020 (1) . Countries that have succeeded in reducing breast cancer mortality have been able to achieve an annual breast cancer mortality reduction of 2–4% per year.
The strategies for improving breast cancer outcomes depend on fundamental health system strengthening to deliver the treatments that are already known to work. These are also important for the management of other cancers and other non-malignant noncommunicable diseases (NCDs). For example, having reliable referral pathways from primary care facilities to district hospitals to dedicated cancer centres.
The establishment of reliable referral pathways from primary care facilities to secondary hospitals to dedicated cancer centres is the same approach as is required for the management of cervical cancer, lung cancer, colorectal cancer and prostate cancer. To that end, breast cancer is a so-called index disease whereby pathways are created that can be followed for the management of other cancers.
Our 24/7 cancer helpline provides information and answers for people dealing with cancer. We can connect you with trained cancer information specialists who will answer questions about a cancer diagnosis and provide guidance and a compassionate ear.
Chat live online
Select the Live Chat button at the bottom of the page
Call us at 1-800-227-2345
Available any time of day or night
Our highly trained specialists are available 24/7 via phone and on weekdays can assist through online chat. We connect patients, caregivers, and family members with essential services and resources at every step of their cancer journey. Ask us how you can get involved and support the fight against cancer. Some of the topics we can assist with include:
For medical questions, we encourage you to review our information with your doctor.
Breast Cancer
It’s important to be able to have frank, open discussions with your cancer care team. They want to answer all of your questions so that you can make informed treatment and life decisions. Here are some questions that you can use to help better understand your cancer and your treatment options.
When deciding on a treatment plan, if you need surgery, during treatment, after treatment, preparing your list of questions.
Once treatment begins, you’ll need to know what to expect and what to look for. Not all of these questions may apply to you, but asking the ones that do may be helpful.
It’s important to be able to have frank, open discussions with your cancer care team. They want to answer all of your questions, so that you can make informed treatment and life decisions.
Not all of these questions will apply to you, but they should help get you started. Be sure to write down some questions of your own. For instance, you might want more information about recovery times or you may want to ask about nearby or online support groups where you can talk with other women going through similar situations. You may also want to ask if you qualify for any clinical trials .
Don’t be afraid to take notes and tell the doctors or nurses when you don’t understand what they’re saying. You might want to bring another person with you when you see your doctor and have them take notes to help you remember what was said.
Keep in mind that doctors aren’t the only ones who can give you information. Other health care professionals, such as nurses and social workers, can answer some of your questions.
To find out more about speaking with your health care team, see The Doctor-Patient Relationship .
The American Cancer Society medical and editorial content team
Our team is made up of doctors and oncology certified nurses with deep knowledge of cancer care as well as editors and translators with extensive experience in medical writing.
Last Revised: November 8, 2021
American Cancer Society medical information is copyrighted material. For reprint requests, please see our Content Usage Policy .
Sign up to stay up-to-date with news, valuable information, and ways to get involved with the American Cancer Society.
If this was helpful, donate to help fund patient support services, research, and cancer content updates.
September 20, 2024 , by Sharon Reynolds
A new study shows how cells in breast tumors (1) recruit nearby nerves to (2) move into the tumors and (3) help the cancer spread to other parts of the body.
The body is packed with sensory nerves that pass sensations like touch, pain, and temperature to the nervous system. Such nerves are tangled through tissues in every organ and many tumors. And according to a new study, breast tumors have an unusual way of exploiting these nerves to help them spread to other parts of the body.
Previous research has found that direct contact between cancer cells and certain kinds of nerve cells can fuel tumor growth. Scientists have even observed some types of cancer cells crawling along nerves to spread ( metastasize ), explained the study’s lead investigator, Veena Padmanaban, Ph.D., a postdoctoral researcher at Rockefeller University.
Although the research team’s work implicated sensory nerves as a critical contributor to breast cancer metastasis, they found that breast cancer cells use them in an entirely different way. The team identified a complex process that starts with blood vessels within tumors releasing a molecule that draws nerves closer to them. The biological changes initiated by this close proximity eventually leads to the activation of metastasis-fueling genes in the cancer cells .
“This is a contact-independent mechanism. Cancer cells don’t have to be touching the nerves for this [communication] to happen,” Dr. Padmanaban explained.
The findings were published August 7 in Nature .
The discovery of this process provides an opportunity for developing treatments that stop it, the study team believes, and potentially prevent breast cancer from spreading. In fact, in the study, they showed that a drug used to prevent nausea and vomiting caused by chemotherapy can block one of the steps of the nerve – cancer cell interaction and stop tumor growth and metastasis in mouse models of breast cancer.
Over the last decade, researchers have begun to uncover the intricate processes by which the nervous system helps tumors grow and spread. The fact that the nervous system appears to contribute to tumor growth isn’t necessarily surprising, said Brunilde Gril, Ph.D., of NCI's Division of Cancer Biology , who was not involved in the study.
“The nervous system is involved in many physiological functions of the body,” Dr. Gril said. For example, nerves support the development of organs and regulate functions of epithelial cells, which are the most abundant cells in the body and give rise to most breast cancers, she explained.
Previous research from the lab of the study’s senior investigator, Sohail Tavazoie, M.D., Ph.D., found that blood vessels in breast tumors that had spread in the body express a protein called SLIT2, which is known to play a role in directing where nerves grow in the body.
In their new study, funded in part by NCI, Dr. Padmanaban and her colleagues from the Tavazoie lab wanted to see if nerves within the tumor microenvironment interacted with breast cancer cells and whether they helped drive tumor growth and spread.
In a series of experiments, they found that blood vessels within tumors had to express SLIT2 to draw in sensory nerves. For example, when they transplanted human breast cancer cells into mice and deleted SLIT2 from endothelial cells , the resulting tumors lost their ability to attract sensory nerves.
Looking at tumor samples collected from people with breast cancer, the team found that people whose breast tumors had a greater abundance of sensory nerves were more likely to have their cancers metastasize. They observed a similar effect in mice implanted with breast cancer cells.
In these experiments, although the sensory nerves appeared to be fueling tumor growth and spread, the two cell types weren’t physically touching, the researchers found. So how were the nerves exerting their effect?
By analyzing and testing substances secreted by both the nerves and the breast cancer cells, the researchers found that a tiny protein called substance P released by sensory nerves substantially boosted growth and spread of breast cancer cells growing in laboratory dishes. Large amounts of substance P were found in samples of human breast tumors that had spread to the lymph nodes.
In mice with breast tumors, blocking substance P greatly reduced tumor growth and metastasis.
But how exactly was substance P promoting metastasis? Dr. Padmanaban and her colleagues found that substance P induces its effects by binding to a receptor on tumor cells called TACR1.
Surprisingly, when this binding occurred, the end result was death for the small subset of breast tumor cells that expressed high levels of the receptor.
When these cells died, they released a type of genetic material called ssRNA, which is normally a signal to the body that a viral infection has occurred.
This ssRNA switched on another receptor found on cancer cells, called TLR7. While TLR7 is involved in the body’s normal immune response , numerous studies have shown that it can also help tumor cells spread.
In this scenario, metastasis “appears to be dependent on a small but significant number of cancer cells dying,” Dr. Tavazoie explained. “But by dying, they’re helping the rest of the cancer cells in the tumor proliferate and metastasize.”
With so many pieces having to fit into place for this process to work, the researchers wondered whether—like pulling out the wrong piece in a Jenga tower—disrupting just one could cause the whole cycle to break down.
Aprepitant, sold under the brand names of Cinvanti and Emend in the United States, is approved to help treat nausea and vomiting caused by some cancer treatments. It works by blocking TACR1, one of the first molecules involved in this newly discovered metastatic process.
In several mouse models of metastatic breast cancer, tumor growth slowed when the researchers treated the mice with aprepitant. And in other mouse models, those that got the drug were much less likely to develop metastases than those that didn’t.
Although aprepitant is an approved drug, it has never been tested in people for potential effects on tumor progression and metastasis, Dr. Gril explained. But the promising results of the experiments in mice suggest that more research to evaluate its potential long-term use and interactions with other cancer treatments are warranted, she added.
The idea of using aprepitant as part of cancer treatment “adds to a bigger conversation that’s been going on about repurposing drugs for cancer,” Dr. Padmanaban said. For example, other researchers recently found that beta blockers prescribed to treat high blood pressure may have an antimetastatic effect. Clinical trials are now testing this idea .
It's not yet clear how early the communication between sensory nerves and cancer cells starts during breast tumor formation, or if other types of cells in the body, such as immune cells, may also play a role in this process, Dr. Padmanaban said, adding “there’s so much more work to be done.”
August 22, 2024, by Carmen Phillips
July 24, 2024, by Sharon Reynolds
July 9, 2024, by Linda Wang
During a mammogram, you stand in front of an X-ray machine designed for mammography. A technician places your breast on a platform and positions the platform to match your height. The technician helps you position your head, arms and torso to allow an unobstructed view of your breast.
Getting a breast MRI involves lying face down on a padded scanning table. The breasts fit into a hollow space in the table. The hollow has coils that get signals from the MRI . The table slides into the large opening of the MRI machine.
A core needle biopsy uses a long, hollow tube to obtain a sample of tissue. Here, a biopsy of a suspicious breast lump is being done. The sample is sent to a lab for testing and evaluation by doctors, called pathologists. They specialize in analyzing blood and body tissue.
Breast cancer diagnosis often begins with an exam and a discussion of your symptoms. Imaging tests can look at the breast tissue for anything that's not typical. To confirm whether there is cancer or not, a sample of tissue is removed from the breast for testing.
During a clinical breast exam, a healthcare professional looks at the breasts for anything that's not typical. This might include changes in the skin or to the nipple. Then the health professional feels the breasts for lumps. The health professional also feels along the collarbones and around the armpits for lumps.
A mammogram is an X-ray of the breast tissue. Mammograms are commonly used to screen for breast cancer. If a screening mammogram finds something concerning, you might have another mammogram to look at the area more closely. This more-detailed mammogram is called a diagnostic mammogram. It's often used to look closely at both breasts.
Ultrasound uses sound waves to make pictures of structures inside the body. A breast ultrasound may give your healthcare team more information about a breast lump. For example, an ultrasound might show whether the lump is a solid mass or a fluid-filled cyst. The healthcare team uses this information to decide what tests you might need next.
MRI machines use a magnetic field and radio waves to create pictures of the inside of the body. A breast MRI can make more-detailed pictures of the breast. Sometimes this method is used to look closely for any other areas of cancer in the affected breast. It also might be used to look for cancer in the other breast. Before a breast MRI , you usually receive an injection of dye. The dye helps the tissue show up better in the images.
A biopsy is a procedure to remove a sample of tissue for testing in a lab. To get the sample, a healthcare professional puts a needle through the skin and into the breast tissue. The health professional guides the needle using images created with X-rays, ultrasound or another type of imaging. Once the needle reaches the right place, the health professional uses the needle to draw out tissue from the breast. Often, a marker is placed in the spot where the tissue sample was removed. The small metal marker will show up on imaging tests. The marker helps your healthcare team monitor the area of concern.
The tissue sample from a biopsy goes to a lab for testing. Tests can show whether the cells in the sample are cancerous. Other tests give information about the type of cancer and how quickly it's growing. Special tests give more details about the cancer cells. For example, tests might look for hormone receptors on the surface of the cells. Your healthcare team uses the results from these tests to make a treatment plan.
Once your healthcare team diagnoses your breast cancer, you may have other tests to figure out the extent of the cancer. This is called the cancer's stage. Your healthcare team uses your cancer's stage to understand your prognosis.
Complete information about your cancer's stage may not be available until after you undergo breast cancer surgery.
Tests and procedures used to stage breast cancer may include:
Not everyone needs all of these tests. Your healthcare team picks the right tests based on your specific situation.
Breast cancer stages range from 0 to 4. A lower number means the cancer is less advanced and more likely to be cured. Stage 0 breast cancer is cancer that is contained within a breast duct. It hasn't broken out to invade the breast tissue yet. As the cancer grows into the breast tissue and gets more advanced, the stages get higher. A stage 4 breast cancer means that the cancer has spread to other parts of the body.
Our caring team of Mayo Clinic experts can help you with your breast cancer-related health concerns Start Here
Breast cancer care at Mayo Clinic
Breast cancer treatment often starts with surgery to remove the cancer. Most people with breast cancer will have other treatments after surgery, such as radiation, chemotherapy and hormone therapy. Some people may have chemotherapy or hormone therapy before surgery. These medicines can help shrink the cancer and make it easier to remove.
Your treatment plan will depend on your particular breast cancer. Your healthcare team considers the stage of the cancer, how quickly it's growing and whether the cancer cells are sensitive to hormones. Your care team also considers your overall health and what you prefer.
There are many options for breast cancer treatment. It can feel overwhelming to consider all the options and make complex decisions about your care. Consider seeking a second opinion from a breast specialist in a breast center or clinic. Talk to breast cancer survivors who have faced the same decision.
A lumpectomy involves removing the cancer and some of the healthy tissue that surrounds it. This illustration shows one possible incision that can be used for this procedure, though your surgeon will determine the approach that's best for your particular situation.
During a total mastectomy, the surgeon removes the breast tissue, nipple, areola and skin. This procedure also is known as a simple mastectomy. Other mastectomy procedures may leave some parts of the breast, such as the skin or the nipple. Surgery to create a new breast is optional. It may be done at the same time as mastectomy surgery or it can be done later.
Sentinel node biopsy identifies the first few lymph nodes into which a tumor drains. The surgeon uses a harmless dye and a weak radioactive solution to locate the sentinel nodes. The nodes are removed and tested for signs of cancer.
Breast cancer surgery typically involves a procedure to remove the breast cancer and a procedure to remove some nearby lymph nodes. Operations used to treat breast cancer include:
Removing the breast cancer. A lumpectomy is surgery to remove the breast cancer and some of the healthy tissue around it. The rest of the breast tissue isn't removed. Other names for this surgery are breast-conserving surgery and wide local excision. Most people who have a lumpectomy also have radiation therapy.
Lumpectomy might be used to remove a small cancer. Sometimes you can have chemotherapy before surgery to shrink the cancer so that lumpectomy is possible.
Removing all of the breast tissue. A mastectomy is surgery to remove all breast tissue from a breast. The most common mastectomy procedure is total mastectomy, also called simple mastectomy. This procedure removes all of the breast, including the lobules, ducts, fatty tissue and some skin, including the nipple and areola.
Mastectomy might be used to remove a large cancer. It also might be needed when there are multiple areas of cancer within one breast. You might have a mastectomy if you can't have or don't want radiation therapy after surgery.
Some newer types of mastectomy procedures might not remove the skin or nipple. For instance, a skin-sparing mastectomy leaves some skin. A nipple-sparing mastectomy leaves the nipple and the skin around it, called the areola. These newer operations can improve the look of the breast after surgery, but they aren't options for everyone.
Complications of breast cancer surgery depend on the procedures you choose. All operations have a risk of pain, bleeding and infection. Removing lymph nodes in the armpit carries a risk of arm swelling, called lymphedema.
You may choose to have breast reconstruction after mastectomy surgery. Breast reconstruction is surgery to restore shape to the breast. Options might include reconstruction with a breast implant or reconstruction using your own tissue. Consider asking your healthcare team for a referral to a plastic surgeon before your breast cancer surgery.
External beam radiation uses high-powered beams of energy to kill cancer cells. Beams of radiation are precisely aimed at the cancer using a machine that moves around your body.
Radiation therapy treats cancer with powerful energy beams. The energy can come from X-rays, protons or other sources.
For breast cancer treatment, the radiation is often external beam radiation. During this type of radiation therapy, you lie on a table while a machine moves around you. The machine directs radiation to precise points on your body. Less often, the radiation can be placed inside the body. This type of radiation is called brachytherapy.
Radiation therapy is often used after surgery. It can kill any cancer cells that might be left after surgery. The radiation lowers the risk of the cancer coming back.
Side effects of radiation therapy include feeling very tired and having a sunburn-like rash where the radiation is aimed. Breast tissue also may look swollen or feel more firm. Rarely, more-serious problems can happen. These include damage to the heart or lungs. Very rarely, a new cancer can grow in the treated area.
Chemotherapy treats cancer with strong medicines. Many chemotherapy medicines exist. Treatment often involves a combination of chemotherapy medicines. Most are given through a vein. Some are available in pill form.
Chemotherapy for breast cancer is often used after surgery. It can kill any cancer cells that might remain and lower the risk of the cancer coming back.
Sometimes chemotherapy is given before surgery. The chemotherapy might shrink the breast cancer so that it's easier to remove. Chemotherapy before surgery also might control cancer that spreads to the lymph nodes. If the lymph nodes no longer show signs of cancer after chemotherapy, surgery to remove many lymph nodes might not be needed. How the cancer responds to chemotherapy before surgery helps the healthcare team make decisions about what treatments might be needed after surgery.
When the cancer spreads to other parts of the body, chemotherapy can help control it. Chemotherapy may relieve symptoms of an advanced cancer, such as pain.
Chemotherapy side effects depend on which medicines you receive. Common side effects include hair loss, nausea, vomiting, feeling very tired and having an increased risk of getting an infection. Rare side effects can include premature menopause and nerve damage. Very rarely, certain chemotherapy medicines can cause blood cell cancer.
Hormone therapy uses medicines to block certain hormones in the body. It's a treatment for breast cancers that are sensitive to the hormones estrogen and progesterone. Healthcare professionals call these cancers estrogen receptor positive and progesterone receptor positive. Cancers that are sensitive to hormones use the hormones as fuel for their growth. Blocking the hormones can cause the cancer cells to shrink or die.
Hormone therapy is often used after surgery and other treatments. It can lower the risk that the cancer will come back.
If the cancer spreads to other parts of the body, hormone therapy can help control it.
Treatments that can be used in hormone therapy include:
Hormone therapy side effects depend on the treatment you receive. The side effects can include hot flashes, night sweats and vaginal dryness. More-serious side effects include a risk of bone thinning and blood clots.
Targeted therapy uses medicines that attack specific chemicals in the cancer cells. By blocking these chemicals, targeted treatments can cause cancer cells to die.
The most common targeted therapy medicines for breast cancer target the protein HER2 . Some breast cancer cells make extra HER2 . This protein helps the cancer cells grow and survive. Targeted therapy medicine attacks the cells that are making extra HER2 and doesn't hurt healthy cells.
Many other targeted therapy medicines exist for treating breast cancer. Your cancer cells may be tested to see whether these medicines might help you.
Targeted therapy medicines can be used before surgery to shrink a breast cancer and make it easier to remove. Some are used after surgery to lower the risk that the cancer will come back. Others are used only when the cancer has spread to other parts of the body.
Immunotherapy is a treatment with medicine that helps the body's immune system to kill cancer cells. The immune system fights off diseases by attacking germs and other cells that shouldn't be in the body. Cancer cells survive by hiding from the immune system. Immunotherapy helps the immune system cells find and kill the cancer cells.
Immunotherapy might be an option for treating triple-negative breast cancer. Triple-negative breast cancer means that the cancer cells don't have receptors for estrogen, progesterone or HER2 .
Palliative care is a special type of healthcare that helps you feel better when you have a serious illness. If you have cancer, palliative care can help relieve pain and other symptoms. A team of healthcare professionals provides palliative care. The team can include doctors, nurses and other specially trained professionals. Their goal is to improve quality of life for you and your family.
Palliative care specialists work with you, your family and your care team to help you feel better. They provide an extra layer of support while you have cancer treatment. You can have palliative care at the same time as strong cancer treatments, such as surgery, chemotherapy or radiation therapy.
When palliative care is used along with all of the other appropriate treatments, people with cancer may feel better and live longer.
Explore Mayo Clinic studies testing new treatments, interventions and tests as a means to prevent, detect, treat or manage this condition.
No alternative medicine treatments have been found to cure breast cancer. But complementary and alternative medicine therapies may help you cope with side effects of treatment.
Many people with breast cancer have fatigue during and after treatment. This feeling of being very tired and worn down can continue for years. When combined with care from your healthcare team, complementary and alternative medicine therapies may help relieve fatigue.
Talk with your healthcare team about:
Some breast cancer survivors say their diagnosis felt overwhelming at first. It can be stressful to feel overwhelmed at the same time you need to make important decisions about your treatment. In time, you'll find ways to cope with your feelings. Until you find what works for you, it might help to:
If you'd like to know more about your breast cancer, ask your healthcare team for the details of your cancer. Write down the type, stage and hormone receptor status. Ask for good sources of information where you can learn more about your treatment options.
Knowing more about your cancer and your options may help you feel more confident when making treatment decisions. Still, some people don't want to know the details of their cancer. If this is how you feel, let your care team know that too.
You may find it helpful and encouraging to talk to others who have been diagnosed with breast cancer. Contact a cancer support organization in your area to find out about support groups near you or online. In the United States, you might start with the American Cancer Society.
Find a friend or family member who is a good listener. Or talk with a clergy member or counselor. Ask your healthcare team for a referral to a counselor or other professional who works with people who have cancer.
Your friends and family can provide a crucial support network for you during your cancer treatment.
As you begin telling people about your breast cancer diagnosis, you'll likely get many offers for help. Think ahead about things you may want help with. Examples include listening when you want to talk or helping you with preparing meals.
Make an appointment with a doctor or other healthcare professional if you have any symptoms that worry you. If an exam or imaging test shows you might have breast cancer, your healthcare team will likely refer you to a specialist.
Specialists who care for people with breast cancer include:
Your time with your healthcare professional is limited. Prepare a list of questions so that you can make the most of your time together. List your questions from most important to least important in case time runs out. For breast cancer, some basic questions to ask include:
In addition to the questions that you've prepared, don't hesitate to ask other questions you think of during your appointment.
Be prepared to answer some questions about your symptoms and your health, such as:
News from mayo clinic.
Mayo Clinic does not endorse companies or products. Advertising revenue supports our not-for-profit mission.
Check out these best-sellers and special offers on books and newsletters from Mayo Clinic Press .
Make your tax-deductible gift and be a part of the cutting-edge research and care that's changing medicine.
Meet CRI’s New CEO Alicia Zhou, PhD
Immune to Cancer: The CRI Blog
Breast cancer is the second most common cancer among U.S. Women. When Breast Cancer Awareness Month was established in 1985, breast cancer was initially resistant to immunotherapy, considered “cold” and unresponsive. Nearly 40 years later, recent advances and clinical trials in immunotherapy are offering new hope, showing potential for better outcomes in breast cancer patients once considered untreatable with these methods.
Immunotherapy changed my life. It saved my life when I had no other options.” Karen Peterson, CRI ImmunoAdvocate and breast cancer survivor
Advocating with Energy and Grace
Thanks to scientific breakthroughs, the five-year survival rate for breast cancer patients has reached 89 percent, and it rises to an incredible 99 percent when the cancer is caught early. This progress has transformed the lives of countless survivors, including Karen Peterson , a CRI ImmunoAdvocate . Karen now dedicates her time to supporting others, sharing her own treatment journey to inspire hope and strength in current patients facing similar challenges. It’s stories like hers that remind CRI just how powerful donor support is in driving life-saving advances.
“I’ve got a second chance to really take care of this body that I’ve worked so hard to keep and stay alive,” Peterson said in conversation with CRI. “I take the responsibility of being an ImmunoAdvocate very seriously.”
Immunotherapy was not her first-line treatment. When Peterson’s cancer returned and spread two years after she was treated with chemotherapy and surgery, she enrolled in a clinical trial. Peterson was eager for treatment and became the first triple-negative breast cancer (TNBC) patient in her immunotherapy trial. Eight weeks later, a CT scan showed that her cancer had shrunk by nearly three-quarters of its original size. TNBC is the most aggressive form of breast cancer, with a higher risk of relapse and metastasis. Unlike other types of breast cancer, TNBC doesn’t rely on hormone signals for its rapid growth, making patients ineligible for hormone therapy. As a result, TNBC patients often face more intensive treatments, combining chemotherapy, radiotherapy, and surgery.
“Standard care didn’t work for me, but everybody’s cancer journey is different,” the Harlem, NY resident explained. She stressed that dealing with cancer involves nuance, and there is no standard rule book. “I feel the renewed energy of patients and caregivers who I speak to about my journey and what it was like to be in a clinical trial. Any emotions, any reactions that you are having are all normal. Give yourself grace and space.”
The Challenges and Opportunities of the Current Breast Cancer IO Landscape
For over 40 years, the Cancer Research Institute (CRI) has been at the forefront of breast cancer immunotherapy, driving innovation and hope. In the 2023-24 fiscal year alone, CRI invested over $4 million in the fight against this devastating disease. CRI-funded scientists are leading groundbreaking research, from tackling TNBC to developing a lactate-responsive drug delivery system that targets cancer metabolism. Their relentless dedication is transforming patient care and bringing us closer to life-saving therapies, offering renewed hope for a future without cancer.
Giulia Furesi , PhD, a CRI Postdoctoral Fellow at Washington University School of Medicine, is researching how a protein in the tumor environment called Osterix impacts the immune system’s ability to fight breast cancer. She explains that breast cancer is difficult to treat because it’s not a one-size-fits-all disease—it’s made up of many different types.
“One significant development has been the growing use of immune checkpoint inhibitors , particularly for TNBC,” she said. Checkpoint inhibitors are an immunotherapy that takes the ‘brakes’ off the immune system, allowing the treatment to target and attack cancer cells. “These inhibitors, which have succeeded in other cancers, are now offering hope for patients with TNBC.”
What’s Next on the Horizon
Sarah Sammons, MD, associate director of the Metastatic Breast Cancer Program at the Dana-Farber Cancer Institute, spoke in detail at CRI’s 2023 Annual Patient Summit about challenges and progress regarding breast cancer immunotherapy. Dr. Sammons noted that immunotherapy has made remarkable progress in the past decade and is now a first-line treatment for some breast cancer subtypes, including TNBC.
“ Pembrolizumab (commonly known by its brand name, Keytruda® ) with five months of chemotherapy before surgery for stage 2 and 3 TNBC has improved cure rates and has improved the pathological complete response,” she said during the patient summit. A pathological complete response means there are no traces of cancer in tissue examined after treatment. “These patients have a 93 to 94 percent chance of being cured of their cancer.”
Dr. Sammons stressed that clinical trials are crucial for discovering new treatments, as they are the pathway for approving drugs for all types of cancer. Currently, there are promising late-stage clinical trials for breast cancer patients, regardless of whether their tumors are PD-L1 positive or negative. PD-L1 is a protein on the surface of tumors that helps them avoid being targeted by the immune system.
Alongside TNBC, immunotherapy can also be effective in treating other breast cancers, as Dr. Sammons explains. “I’m very hopeful that patients with fast-growing hormone receptor-positive breast cancer may have an opportunity to get immunotherapy in the future. This is on the horizon.” Hormone receptors are proteins located on breast cancer cells, and they can latch on to estrogen or progesterone signals, promoting cancer cell growth. Eligible patients with tumors expressing these hormone receptors can be treated by using hormone antagonist drugs. With advancements in immunotherapy these patients will have more options of treatment available for them.
Thanks to the tireless research of scientists like Drs. Furesi and Sammons, and the dedicated advocacy of passionate breast cancer survivors like Karen Peterson, we are closer to creating a world immune to cancer.
Live from new york.
Hosted by Cecily Strong Actress and Comedian
Annual Awards Gala October 15, 2024
Cipriani 25 Broadway, New York City
This website uses tracking technologies, such as cookies, to provide a better user experience. If you continue to use this site, then you acknowledge our use of tracking technologies. For additional information, review our Privacy Policy .
We asked Dr. Graham King, a high-risk breast consultant, to share the top 10 questions about breast cancer that he hears from patients.
Breast cancer receives much attention during October’s awareness month. Although sightings of pink ribbons and breast cancer-related information increase during the month, it’s crucial to keep screenings and clinical breast exams at the forefront of preventive care year-round.
We asked Dr. Graham King, a high-risk breast consultant and breast cancer awareness advocate with the Mayo Clinic Health System, to share the top 10 questions about breast cancer that he hears from patients.
Why do I need to worry about breast cancer and having routine mammograms?
This is the most common question shared during breast clinic consultations. Many people mention that they don’t have any family history of breast cancer. However, approximately just 20 percent of breast cancer is related to family history and genetic links. People are considered at the highest risk after a breast cancer diagnosis in a first-degree relative, such as a mom or sister.
What can I do to prevent breast cancer from developing?
For most people, the answer is to live a healthy lifestyle, including not smoking, minimizing alcohol consumption and maintaining an ideal body weight through diet and exercise. The majority of the risk for breast cancer comes from being female, having breasts and aging.
What steps can I take to be informed of my risk of breast cancer?
This answer has two parts. The first part is to learn about your family’s medical history. The second part is to follow your health care team’s recommended preventive screening plan, including a mammogram if necessary.
Should I continue doing self-checks of my breasts and have an annual clinical breast exam?
The American College of Obstetricians and Gynecologists and the U.S. Preventive Services Taskforce update breast screening recommendations annually, but more research studies need to be done. Your health history and conversations with your health care team will help inform the best approach for you. Although not all health care professionals perform a yearly clinical breast exam as part of an annual physical, you may choose to perform monthly or quarterly self-checks to increase your breast awareness. Both a yearly breast exam by a medical professional and regular self-checks are recommended if you have a higher risk of breast cancer.
What does it mean to have dense breasts, and how does that affect mammograms?
Approximately 30 percent of people have moderately dense breasts, and up to 10 percent have extremely dense breasts. While dense breast tissue does affect the detection of breast cancer through mammography, a mammogram is still a recommended annual screening starting at age 40 for people with average breast cancer risk.
Does taking a birth control pill increase my risk of developing breast cancer?
No strong connection has been identified to suggest any such connection in average-risk patients during childbearing years. However, studies suggest that continuing hormone therapy after age 60 can increase the risk of breast and endometrial cancer.
Does pregnancy and breastfeeding increase my risk of breast cancer?
No, it’s quite the opposite. The risk of developing breast cancer decreases based on the duration of time spent pregnant and breastfeeding.
How do environmental toxin exposure and radiation affect breast cancer?
Certain factors, such as radiation exposure from previous cancer treatment, working in an environment with toxins, or other radiation exposure can increase your risk of many cancer types, including breast cancer.
Should I have genetic testing to determine if I have a family-related risk of breast cancer?
The short answer is, for some people, yes. However, genetic testing is recommended only after a discussion with your health care team or breast specialist about your cancer risks. You may be asked to meet with a genetics counselor to ensure that there is a strong indication for testing.
Does my race affect my risk of breast cancer?
The answer to this question is complex. According to the Centers for Disease Control and Prevention, African Americans, American Indians, Pacific Islanders and Alaskan Native Americans have a much higher rate of breast cancer and breast cancer-related mortality. However, there are many nuances to this concerning medical equity, racial disparity and other considerations that need to be explored and addressed.
If you have been identified as having a higher risk of breast cancer and referred to a high-risk breast clinic, you can expect a comprehensive meeting with a health care professional who is skilled and passionate about breast cancer prevention and survivorship.
Michele Klimke has been a dedicated medical dosimetrist for forty years and joined the Comprehensive Cancer Centers of Nevada (Comprehensive) team in 2007. With extensive experience in developing and managing radiation plans for patients undergoing cancer treatment, she never anticipated finding herself on the other side of the machine. After undergoing a routine mammogram, Michele […]
In our daily lives, twelve feet is a familiar measure – the height of an extension ladder, the top of a basketball backboard, and the width of a lane on freeways across America. However, when it comes to an emergency responder or a driver in distress on the side of the road, twelve feet can […]
Dental implants are quickly becoming the preferred solution for replacing missing teeth, whether you need to replace one, several, or all your teeth, depending on the condition of your oral health. Implants are known for their durability, functionality, and natural appearance. In this blog, guided by tips from 4M Dental Implant Centers, leaders in the […]
Did you know? The updated flu and COVID-19 vaccinations for 2024-2025 are now available at Southern Nevada Health District clinics. You can receive both vaccines at the same time. The Health District and the Centers for Disease Control and Prevention (CDC) recommend that everyone eligible get their updated vaccinations to help protect against severe illness […]
Historic numbers of Americans are living on their own as they age, and it can have profound health consequences.
Experts tell us that a robust immune system is one of the most potent weapons we have against viral illnesses such as influenza and COVID.
The Aging Wellness Expo is more than just an event — it’s an opportunity to shape a more inclusive, age-positive future.
“I don’t know what to expect when I step onto a new set. But I choose to turn that fear into excitement,” the two-time Oscar winner says.
SSI, or supplemental security income, provides monthly cash benefits, based on financial need, to people who are disabled or over age 65.
People need to understand the differences between original Medicare with a supplement and Medicare Advantage PPO plans.
Many people consider eating less meat. But they sometimes hesitate because they are concerned about how they would replace all that lost protein.
More than a million people will probably have to find new coverage as major insurers cut costs and pull back from markets for Medicare Advantage plans.
Arguably the most talented therapist I know, Las Vegan Jimmy Monaghan has lived a life that could well inspire a smash-hit Netflix documentary.
Choosing which surgical route to take is deeply personal, and there is no one “best choice” for every woman.
For its influential Stop Breast Cancer for Life public service campaign supporting BCRF, longtime partner Lifetime has teamed up with stars Lyndsy Fonesca, Karrueche Tran, and Jackie Cruz to highlight rising rates of breast cancer in younger women and the importance of early detection and learning your personal risk .
“We are grateful for Lifetime’s continued support in raising awareness about breast cancer and advancing our mission,” BCRF’s Chief Scientific Officer Dorraya El-Ashry said. “While the incidence of breast cancer in younger women remains low, diagnoses in women under 50 are rising. We’re making strides in understanding the disease better and improving care for younger patients, with the goal of reversing this trend. Lifetime’s ongoing commitment to highlighting breast cancer year after year plays a vital role in propelling this lifesaving research forward.”
This marks the 30 th year of Lifetime’s Stop Breast Cancer for Life initiative. The PSA campaign will air on Lifetime and across digital platforms and will be amplified by Lifetime’s distribution partners. Stop Breast Cancer for Life will also feature four Lifetime films with breast cancer at the center including the breast cancer short film anthology FIVE from directors Jennifer Aniston, Demi Moore, Alicia Keyes, Penelope Spheeris, and Patty Jenkins and starring Fonseca, Jeanne Tripplehorn, and Patricia Clarkson. "Breast cancer continues to affect far too many lives, including women of all ages and backgrounds,” A+E Networks Group President and Chairman Paul Buccieri said. “A+E Networks is proud to join BCRF to raise awareness about early detection, galvanize funding for critical research, and continue to pave the way to find a cure and breast cancer once and for all.”
Watch the PSA above and learn more about Stop Breast Cancer for Life here .
Connect with us.
Please remember BCRF in your will planning. Learn More
Breast Cancer Research Foundation 28 West 44th Street, Suite 609, New York, NY 10036
General Office: 646-497-2600 | Toll Free: 1-866-346-3228 [email protected] | BCRF is a 501 (c)(3) | EIN: 13-3727250
From your first visit to after diagnosis and treatment
If you’ve been told you have breast cancer , it’s important to ask your doctor and other healthcare providers questions so you thoroughly understand your diagnosis , treatment plan options, and what you can expect throughout treatment and recovery.
Because this can be an overwhelming time, it’s a good idea to write down any questions you have in advance and bring them with you to your medical appointments. Bring a notepad so you can write down the answers to your questions. If possible, bring a family member or friend with you to appointments as well.
Srdjanns74 / Getty Images
This article will go over questions to ask after you initially receive a breast cancer diagnosis, before, during, and after treatment, before and after surgery, and when all treatments are completed.
A cancer care team is made up of different types of healthcare providers. In addition to an oncologist , providers you might see if you have breast cancer include:
Other members of your care team can include physician assistants, nurse practitioners, nurses, mental health professionals, nutritionists, social workers, and patient/nurse navigators.
If you’ve been diagnosed with breast cancer, you’ll want to learn more about your type of breast cancer , treatment options, and support. If your primary healthcare provider suspects you might have cancer, they will likely refer you to an oncologist (a doctor specializing in diagnosing and treating cancer).
The following are initial questions to ask your oncologist:
Genetic testing can help determine if your breast cancer or family history of breast cancer is due to an inherited gene mutation, which can help guide treatment. Only 5–10% of breast cancers are related to an inherited gene mutation. Questions to ask your doctor about genetic testing include:
Treatments for breast cancer include surgery , radiation , chemotherapy , hormone therapy , targeted therapy, and immunotherapy . Treatments can be given separately or combined. Your treatment options will depend on your type of cancer, its stage, your health, and other factors. It's important to ask questions to your radiation oncologist, medical oncologist, and surgical oncologist or breast surgeon before, during, and after treatment.
Questions to ask before treatment include:
Questions to ask during treatment include;
Questions to ask after treatment include:
Make sure to ask your healthcare providers how you can communicate or ask questions outside of scheduled appointment times. Find out if there are options such as email, patient portals, or after-hours phone services available and how you can access them.
Surgery is a part of breast cancer treatment for most people. Types of surgeries include breast-conserving surgery (lumpectomy) to remove the tumor and surrounding tissue, mastectomy to remove the entire breast, breast reconstruction surgery , and procedures to remove lymph nodes .
If you’re having surgery, there are specific questions you should ask your breast surgeon or surgical oncologist before and after your surgery.
Questions to ask before surgery include:
A patient advocate is an important resource for assisting you with healthcare decisions, problems, coordination of care, and more. Different types of patient advocates include those affiliated with nonprofit organizations, for-profit organizations, or hospitals. In addition, private patient advocates can be hired on an individual basis.
Questions to ask after surgery include:
When you complete your treatment, you will still need to make follow-up appointments. You can ask any doctors or other healthcare providers who are involved in your follow-up care the following questions:
Though a breast cancer diagnosis can be overwhelming, it's important to ask questions throughout every stage of your care. It's a good idea to write down questions in advance of your medical appointments. Make sure you have a list of questions to ask after your initial diagnosis, throughout treatment, before and after surgery, and when your treatments are complete.
Remember, you are the most important member of your healthcare team. Don’t ever be afraid to ask your doctors and other healthcare providers questions about your diagnosis, treatment, recovery, or anything else that concerns you. Doing so will help you make informed decisions about your cancer care.
For your first visit, you can narrow questions down to those about the type of cancer you have, treatment options, and prognosis. You can also ask your doctor how to contact them if you have further questions after your first appointment.
Cancer stage will be determined by how large your tumor is, whether it has hormone receptors, and whether it has spread. It is given a number between 0 and 4. A biopsy (microscopic examination of a sample of the tumor) first confirms you have breast cancer. A clinical stage may be assigned before surgery, and this may be updated (pathological or surgical stage) after surgery and further imaging or tests.
Additional questions to ask if you have been diagnosed with invasive breast cancer include:
American Cancer Society. Treating breast cancer .
Lindenberg Cancer & Hematology Center. When are patients referred to an oncologist?
Susan G. Komen. Genetic testing after a breast cancer diagnosis .
Susan G. Komen. Genetic testing to learn about breast cancer risk .
American Cancer Society. Surgery for breast cancer .
Breastcancer.org. Breast cancer stages .
American Society of Clinical Oncology. Breast cancer-metastatic: questions to ask the health care team .
By Cathy Nelson Nelson is a freelance writer specializing in health, wellness, and fitness for more than two decades.
A new U.S. rule requires mammography centers to inform women about their breast density
When a woman has a mammogram, the most important finding is whether there’s any sign of breast cancer.
The second most important finding is whether her breasts are dense.
Since early September, a new U.S. rule requires mammography centers to inform women about their breast density — information that isn’t entirely new for some women because many states already had similar requirements.
Here’s what to know about why breast density is important.
No, dense breasts are not bad. In fact, they’re quite normal. About 40% of women ages 40 and older have dense breasts.
Women of all shapes and sizes can have dense breasts. It has nothing to do with breast firmness. And it only matters in the world of breast cancer screening, said Dr. Ethan Cohen of MD Anderson Cancer Center in Houston.
With the new rule, “there are going to be a lot of questions to a lot of doctors and there’s going to be a lot of Googling, which is OK. But we want to make sure that people don’t panic,” Cohen said.
Doctors who review mammograms have a system for classifying breast density.
There are four categories. The least dense category means the breasts are almost all fatty tissue. The most dense category means the breasts are mostly glandular and fibrous tissue.
Breasts are considered dense in two of the four categories: “heterogeneously dense” or “extremely dense.” The other two categories are considered not dense.
Dr. Brian Dontchos of the Seattle-based Fred Hutchinson Cancer Center said the classification can vary depending on the doctor reading the mammogram "because it’s somewhat subjective.”
Two reasons: For one, dense breasts make it more difficult to see cancer on an X-ray image, which is what a mammogram is.
“The dense tissue looks white on a mammogram and cancer also looks white on a mammogram,” said Dr. Wendie Berg of the University of Pittsburgh School of Medicine and chief scientific adviser to DenseBreast-info.org. “It’s like trying to see a snowball in a blizzard.”
Second, women with dense breast tissue are at a slightly higher risk of developing breast cancer because cancers are more likely to arise in glandular and fibrous tissue.
Reassuringly, women with dense breasts are no more likely to die from breast cancer compared to other women.
If you find out you have dense breasts, talk to your doctor about your family history of breast cancer and whether you should have additional screening with ultrasound or MRI, said Dr. Georgia Spear of Endeavor Health/NorthShore University Health System in the Chicago area.
Researchers are studying better ways to detect cancer in women with dense breasts. So far, there’s not enough evidence for a broad recommendation for additional screening. The U.S. Preventive Services Task Force called for more research in this area when it updated its breast cancer screening recommendations earlier this year.
Yes, women with dense breasts should get regular mammograms, which is still the gold standard for finding cancer early. Age 40 is when mammograms should start for women, transgender men and nonbinary people at average risk.
“We don’t want to replace the mammogram,” Spear said. “We want to add to it by adding a specific other test.”
For now, that depends on your insurance, although a bill has been introduced in Congress to require insurers to cover additional screening for women with dense breasts.
Additional screening can be expensive — from $250 to $1,000 out of pocket, so that’s a barrier for many women.
“Every woman should have equal opportunity to have their cancer found early when it’s easily treated,” Berg said. “That’s the bottom line.”
The Associated Press Health and Science Department receives support from the Howard Hughes Medical Institute’s Science and Educational Media Group. The AP is solely responsible for all content.
24/7 coverage of breaking news and live events
Together we are beating cancer
The latest news, analysis and opinion from Cancer Research UK
9 October 2024
This month, we’ve been marking the 30 th anniversary of the discovery of the BRCA1 gene . That discovery, along with that of BRCA2 just a year later, revolutionised how we prevent, diagnose and treat certain cancers.
Faulty BRCA genes can be inherited from either parent, and people with these mutations can have an increased risk of developing certain cancers, including breast, ovarian, prostate and pancreatic cancer.
Here, three women with faulty BRCA genes share their experiences of cancer.
Maria’s experience of cancer started at an early age, when her mum lost two of her sisters, both in their 30s, to breast cancer. Her mum’s grandmother also died from the disease.
“Fast forward 30 years to my own grandmother and her two sisters passing away from the same thing. It was obviously a worry in the family.
“Surely that can’t be coincidence; something about that screams it must be more than just bad luck – it must be genetic.”
Then, in 2018, Maria’s mum was also diagnosed with breast cancer and tests showed that she had the BRCA2 gene mutation.
This resulted in the rest of Maria’s family also coming forward to have genetic testing. They were also eligible for earlier breast cancer screening from the age of 30, so Maria booked a mammogram while she was waiting for her genetic test results to come through.
Unfortunately, the mammogram showed that she, like her mum, had breast cancer.
“It was such a blow, as my husband and I were planning to get married and try for a baby and were only able to have one round of IVF before I started chemotherapy.”
Maria’s first treatment was a lumpectomy in October 2018 and shortly after, her genetic test results came back showing that she also had the BRCA2 gene mutation.
Maria had six cycles of chemotherapy followed by a double mastectomy with simultaneous reconstruction with implants.
Maria is now on a five-year course of tamoxifen, a hormone therapy that can help to reduce the risk of breast cancer coming back.
She’s also waiting for a preventative oophorectomy (surgical removal of the ovaries) as the BRCA2 gene also increases the risk of ovarian cancer.
Around 8 in every 10 breast cancers diagnosed in the UK are classified as oestrogen receptor-positive, or ER-positive for short. This means that they are encouraged to grow by the hormone oestrogen, which circulates in the bloodstream. Tamoxifen stops oestrogen from affecting cancer cells, stopping them growing.
We supported clinical trials showing that taking tamoxifen could help prevent breast cancer in certain women who are at higher risk of the disease.
Our researchers also helped prove the benefits of taking tamoxifen after surgery, for women with the most common type of breast cancer.
Learn more about our work into tamoxifen .
“I have been in early menopause since the end of my treatment, which has left me feeling drained, tired and in pain. Everyone thinks you just get back to normal after treatment for cancer, but I have found everything a struggle, including just going to work every day.
“It has been particularly hard coming to terms with the fact I will never be able to have children, but I am grateful every day to be here.”
Maria’s big family meant many of her relatives were eligible for genetic testing.
“My mother is 1 of 10 children and I have countless first and second cousins. I am one of six children of whom four tested positive, as did several of my uncles and aunties; nine of us in all have had preventative surgery, along with those who have had surgery due to having cancer.
“Had my mum not had the genetic test I might not be here now. Instead, I have survived for more than five years after diagnosis and been treated, as has my mum. We are all so thankful to her; the best way I can show this is by telling our story and helping to spread awareness of the fact that there can sometimes be a genetic link to cancer.”
At the age of 38, almost a year after losing her mum to breast cancer in June 1999, Gillian found a small lump in her breast. She visited her GP who sent her to the local hospital to get it checked.
After a needle biopsy, where a sample of cells is taken using a small needle, and an ultrasound, Gillian was diagnosed with a rare type of cancer called medullary breast cancer .
Medullary breast cancer occurs more often in younger women and in women who have inherited a faulty BRCA1 gene.
Fortunately, as the cancer had been caught early, it hadn’t spread. So, Gillian had surgery to remove the lump and surrounding tissue. Her doctors also recommended five weeks of radiotherapy once she had recovered from her operation.
As part of her treatment, Gillian took part in the Cancer Research UK-funded START trial.
“The treatment wasn’t too bad, although I did get very tired. I followed the nurses’ advice and although it was a bit uncomfortable the area being treated wasn’t painful.”
We funded the START trial, which was split into two branches, START A and START B . The trials looked at different ways of giving radiotherapy after surgery for early-stage breast cancer.
In 2013 the researchers published their 10-year follow up of the trials. This showed that fewer, but higher, doses of radiotherapy is as effective as the then-standard dose of 50 Gy in 25 fractions.
As a result, most UK centres have now adopted a dose schedule of 40 Gy in 15 fractions as the standard care for women with breast cancer.
“Then in March 2007 I started to feel really bloated which was unusual for me as I’d always been rather slim. I wondered if it was something to do with my menstrual cycle or maybe menopause. But my GP said it wasn’t normal for me and sent me straight off for a blood test and ultrasound.”
The ultrasound showed a solid mass on one side of her stomach and a fluid filled mass on the other side. Gillian was referred for a CT scan and further blood tests, which showed that she had ovarian cancer.
“By this time, I looked like I was six months pregnant and on 7 June 2007 I had a full hysterectomy, and the surgeons removed a tumour the size of a football.
“A few weeks after the operation I started chemotherapy – a combination of carboplatin and paclitaxel. Chemo wasn’t as bad as I’d thought it would be. The worst side effect was that I ached all over, it was hard to sleep I ached so much. Losing my hair was no big deal really; it was a case of saving my life or saving my hair, so it was an easy decision to make!”
Gillian had five cycles of chemotherapy, finishing in November 2007.
Then, a month later in December, Gillian tested positive for the BRCA1 gene mutation, which she was found to have inherited from her father.
Because this meant that she had an increased risk of her breast cancer returning, Gillian had a double mastectomy with implant reconstruction in March 2023.
She has since made a good recovery from the surgery and is doing well.
Sue first started noticing signs that something wasn’t quite right for her body in November 2016 when she started gaining weight, despite trying to lose it.
“Over Christmas I felt bloated but put it down to overindulgence until my sister came to stay. We talked about our recent discovery that our cousin, who had had breast cancer, had been found to carry a BRCA gene mutation.
“Our mother had died of breast cancer, but although we had not been tested my sister drew my attention to the fact the gene could also be associated with ovarian cancer.”
On the advice of her sister, Sue made a GP appointment, where she was referred for an ultrasound and blood tests. The next day, her GP called her in urgently to tell her that her levels of CA125 – a protein that is often higher in people with ovarian cancer – were very high. The ultrasound found a 15cm mass, further pointing towards a suspected ovarian cancer diagnosis.
“At the end of January, the consultant confirmed the diagnosis of ovarian cancer. I was with my sister, who is a nurse and was able to take in all the information I was given.
“Trying to remain strong whilst all I wanted to do was scream was a real challenge. If it wasn’t for my husband, I would have fallen apart; he was and is still my rock and an absolute hero.”
Sue had a hysterectomy in February 2017 and started chemotherapy in March as part of a clinical trial called JAVELIN Ovarian.
During her chemotherapy, Sue was tested for BRCA gene mutations and found out she had a faulty BRCA1 gene.
As she had been part of a trial, Sue had regular CT scans and blood tests. Unfortunately, in June 2018 there was a rise in her CA125 levels. By the autumn her CA125 had increased even more, and she was put back on chemotherapy.
In March 2019, Sue joined the Cancer Research UK funded ICON9 trial . This trial was looking at the effectiveness of long-term treatment with two drugs called olaparib and cediranib for women whose ovarian cancer has started to grow again.
Sue was in the group taking just olaparib tablets twice daily.
“I feel so lucky my oncologists are the chief investigators of the trial. I am currently on two-monthly investigations, and my CA125 count has remained stable.
Olaparib, which is one of the drugs that was used in the ICON9 trial, belongs to a group of drugs known as PARP inhibitors.
PARP inhibitors block a protein called PARP that helps damaged cancer cells to repair themselves. By blocking PARP, this helps stop cancer cells repairing themselves.
Our researchers led the underpinning work to understand PARP’s role in DNA repair, and we supported the early development and first-in-human testing of two early PARP inhibitor drugs.
Learn more about how we were involved in the development of PARP inhibitors .
Sue, who has three children, is now doing well.
“My health in general is great and all my children treat me as if life is back to normal, which is lovely,” said Sue.
“As someone who is receiving first-hand the benefits of the wonderful research of Cancer Research UK, I am endlessly thankful and fully appreciate the relentless funding that is needed to achieve so many new therapies and wonderful outcomes. Without funding and the tireless work of Cancer Research UK, we would have no success stories to share.”
Since the discovery of the BRCA genes 30 years ago, we’ve made huge leaps forward in understanding faulty BRCA-driven cancers – leaps that are saving and improving lives right now.
But our work isn’t done yet. We want to bring about a world where everybody lives longer, better lives free from the fear of cancer. No matter who they are or where they’re from.
Absolutely brilliant, this is why I donate, for people who are sick with this terrible disease, to help towards getting better treatments for people who are trying to fight this disease.
Thank you for the BRCA information. I have recently been diagnosed with BRCA 1 and had a double mastectomy and awaiting removal of ovaries and fallopian tubes.
Leave a reply cancel reply.
Your email address will not be published. Required fields are marked *
Save my name, email, and website in this browser for the next time I comment.
Read our comment policy .
30 years of brca: how the discovery of two breast cancer genes continues to drive progress, cancer metabolism: finding how fast-growing cancers get their energy, groundbreaking trial launches for everyone with ewing sarcoma, introducing diy steps to genetic testing could catch more brca-linked cancer cases, we’re accelerating brain tumour research with £8m more for our brain tumour centres of excellence, cancer vaccines - where are we, more like this, olaparib approved for 800 prostate and breast cancer patients in england, how serendipity sent this oncologist in search of a cure for cancer, related topics.
I had a primary breast cancer, had a lumpectomy and lymph nodes removed. No cancer found in the nodes. My question. If it came back in the same breast or the other. Is that described as terminal..?
Hello and thanks for posting
If breast cancer comes back in the same breast it is called a local recurrence. If it appears in the other breast then this would be classified as a new primary breast cancer. This can often be treated effectively with further surgery, radiotherapy or other treatments as recommended by the breast care team.
It sounds from what you have described that you had an early cancer which has not spread to nearby lymph nodes and as a result is less likely to spread in the future.
Your breast care team or nurse specialist can advise you further on this as they are familiar with all your medical details.
I hope this is of some help. Give us a ring if you would like to talk anything over. The number to call is Freephone 0808 800 4040 and the lines are open from 9am till 5pm Monday to Friday.
Kind regards,
Hi Vivienbegonia,
A very warm welcome to our forum.
I was diagnosed with a primary cancer in one breast and had a lumpectomy. There was no lymph node involvement. Less than a year later I discovered another lump, in the same breast. This was also cancerous. This was a second primary cancer. I had a double mastectomy and I still lead a busy and fulfilling life, even though that was all 14 years ago now.
Jolamine xx
Thank you Jolamine, so happy for you.
No problem, Vivienbegonia. Have you had a recurrence?
Hi Jolamine
No I have not had a recurrence. Xx
Good, I'm glad to hear that you haven't had a recurrence.
Number | Subject | Description | Edited User Id |
---|---|---|---|
Current | Secondary breast can… | I had a primary breast cancer, had a lumpectomy and… | Vivienbegonia |
IMAGES
VIDEO
COMMENTS
What were the top 10 review topics? Rank. Topic in Cochrane format. Topic question in lay terms. 1. Omission of whole breast irradiation for postmenopausal women with early breast cancer. Can radiotherapy be safely omitted in postmenopausal women with early breast cancer? 2. Platinum-containing regimens for neoadjuvant and adjuvant therapy in ...
Some research is basic, exploring questions as diverse as the biological underpinnings of cancer and the social factors that affect cancer risk. And some are more clinical, seeking to translate this basic information into improving patient outcomes. ... Breast Cancer Research Results. The following are some of our latest news articles on breast ...
Breast cancer is currently one of the most prevalently diagnosed cancers and the 5th cause of cancer-related deaths with an estimated number of 2.3 million new cases ... the key question is the discrimination between patients who will or will not benefit from particular therapies. ... International Agency for Research on Cancer; Lyon, France ...
Posted: January 20, 2023. Many young women who are diagnosed with early-stage breast cancer want to become pregnant in the future. New research suggests that these women may be able to pause their hormone therapy for up to 2 years as they try to get pregnant without raising the risk of a recurrence in the short term.
Introduction. Breast cancer is the most commonly diagnosed cancer among female patients and is the leading cause of cancer-related death. 1 There were 300,590 new cases and 43,700 deaths of invasive breast cancer in the United States based on the 2023 prediction, accounting for approximately 30% of female cancers. 1 The treatments of breast cancer include surgery, chemotherapy, radiotherapy ...
Breast cancer (BC) is a highly prevalent malignancy worldwide, with complex pathogenesis and treatment challenges. Research reveals that methyltransferase-like 3 (METTL3) is widely involved in the pathogenesis... Dongqiong Xiao, Mingfu Zhang, Yi Qu and Xiaojuan Su. Breast Cancer Research 2024 26:110.
New research questions emerge as medical needs continue to evolve and as we improve our understanding of cancer biology and treatment of malignancies. Although significant advances have been made in some areas of breast cancer research resulting in improvements in therapies and outcomes over the last few decades, other areas have not benefited to the same degree and we continue to have many ...
Breast cancer (BC) is the most frequent cancer diagnosed in women worldwide. This heterogeneous disease can be classified into four molecular subtypes (luminal A, luminal B, HER2 and triple-negative breast cancer (TNBC)) according to the expression of the estrogen receptor (ER) and the progesterone receptor (PR), and the overexpression of the human epidermal growth factor receptor 2 (HER2).
Breast Cancer Research is an international, peer-reviewed online journal, publishing original research, reviews, editorials and reports. Open access research articles of exceptional interest are published in all areas of biology and medicine relevant to breast cancer, including normal mammary gland biology, with special emphasis on the genetic, biochemical, and cellular basis of breast cancer.
We conducted a survey with input from breast cancer physicians, laboratory-based researchers, and patients to analyze the current understanding of ILC, and identify consensus research questions ...
Impressive gains in breast cancer research and treatment have been made over the past 45 years in high-income countries. Many women are now cured with a simple lumpectomy, minimal lymph node surgery, and targeted or endocrine therapy. 1 However, this progress is in stark contrast to what exists in low- and middle-income countries (LMICs) 2 where women have a higher burden of breast cancer ...
Possible environmental causes of breast cancer have also received more attention in recent years. While much of the science on this topic is still in its earliest stages, this is an area of active research. Breast cancer prevention. Researchers are looking for ways to help reduce breast cancer risk, especially for women who are at high risk.
Eye-Opening Breast Cancer Statistics on Racial Disparities. 0.5%. average increase in breast cancer incidence rates each year since the mid-2000s. 200%. increased likelihood Black women have of developing triple-negative breast cancer. 41%. increase in the breast cancer death rate for Black women compared with White women.
Since the beginning of 2017, Cancer Communications (former title: Chinese Journal of Cancer) has published a series of important questions regarding cancer research and clinical oncology, to provide an enhanced stimulus for cancer research, and to accelerate collaborations between institutions and investigators. In this edition, the following 8 valuable questions are presented.
The average age of Black women diagnosed with breast cancer is 60 years old, compared to an average age of 62 for white women. 1. Black women are 40% more likely to die from breast cancer than white women. 1. Black women have the lowest 5-year relative breast cancer survival rate of any racial or ethnic group. 1.
Treatment for inflammatory breast cancer and overall survival. CDK4/6 inhibitors and treatment for metastatic breast cancer. Elacestrant (Orserdu) and treatment for metastatic breast cancer. PI3 kinase inhibitors and AKT inhibitors for metastatic breast cancer treatment. Trastuzumab (Herceptin) and treatment for metastatic breast cancer.
Breast cancer is a disease in which abnormal breast cells grow out of control and form tumours. If left unchecked, the tumours can spread throughout the body and become fatal. Breast cancer cells begin inside the milk ducts and/or the milk-producing lobules of the breast. The earliest form (in situ) is not life-threatening and can be detected ...
Questions to Ask Your Doctor About Breast Cancer. It's important to be able to have frank, open discussions with your cancer care team. They want to answer all of your questions so that you can make informed treatment and life decisions. Here are some questions that you can use to help better understand your cancer and your treatment options.
A Mayo Clinic Health System high-risk breast consultant and breast cancer awareness advocate shares the top 10 questions about breast cancer asked by people of all ages and backgrounds. ... but more research studies need to be done. Your health history and conversations with your healthcare team will help inform the best approach for you.
Breast cancer questions are common among patients diagnosed with the condition. Discover the answers to your most common breast cancer questions. We're available 24/7. Call us anytime. (888) 552-6760 (888) 552-6760 CHAT NOW SCHEDULE AN APPOINTMENT. How we treat cancer. Cancers We Treat. Breast Cancer;
Although the research team's work implicated sensory nerves as a critical contributor to breast cancer metastasis, they found that breast cancer cells use them in an entirely different way. The team identified a complex process that starts with blood vessels within tumors releasing a molecule that draws nerves closer to them.
Mammogram. A mammogram is an X-ray of the breast tissue. Mammograms are commonly used to screen for breast cancer. If a screening mammogram finds something concerning, you might have another mammogram to look at the area more closely. This more-detailed mammogram is called a diagnostic mammogram.
The Challenges and Opportunities of the Current Breast Cancer IO Landscape. For over 40 years, the Cancer Research Institute (CRI) has been at the forefront of breast cancer immunotherapy, driving innovation and hope. In the 2023-24 fiscal year alone, CRI invested over $4 million in the fight against this devastating disease.
We asked Dr. Graham King, a high-risk breast consultant and breast cancer awareness advocate with the Mayo Clinic Health System, to share the top 10 questions about breast cancer that he hears ...
Lifetime's ongoing commitment to highlighting breast cancer year after year plays a vital role in propelling this lifesaving research forward." This marks the 30 th year of Lifetime's Stop Breast Cancer for Life initiative. The PSA campaign will air on Lifetime and across digital platforms and will be amplified by Lifetime's ...
Genetic testing can help determine if your breast cancer or family history of breast cancer is due to an inherited gene mutation, which can help guide treatment. Only 5-10% of breast cancers are related to an inherited gene mutation. Questions to ask your doctor about genetic testing include: Should I consider genetic testing or see a genetic ...
Based on the high demand for copper in breast cancer, we prepare a fibrous therapeutic nanoagent (Zn-PEN) by chelating D-Penicillamine (D-PEN) with Zn2+. Firstly, Zn-PEN achieves dual inhibition of oxidative phosphorylation and glycolysis metabolism through copper depletion and Zn2+ activated cGAS-STING pathway, thus inducing tumor cell death.
A new U.S. rule requires mammography centers to inform women about their breast density When a woman has a mammogram, the most important finding is whether there's any sign of breast cancer. The ...
Medullary breast cancer occurs more often in younger women and in women who have inherited a faulty BRCA1 gene. Fortunately, as the cancer had been caught early, it hadn't spread. So, Gillian had surgery to remove the lump and surrounding tissue. Her doctors also recommended five weeks of radiotherapy once she had recovered from her operation ...
I had a primary breast cancer, had a lumpectomy and lymph nodes removed. ... No cancer found in the nodes. My question. If it came back in the same breast or the other. Cancer Research UK main site. ... Cancer Research UK is a registered charity in England and Wales (1089464), Scotland (SC041666), the Isle of Man (1103) and Jersey (247). ...