Review Open Access
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Apr 24, 2020; 11(4): 169-179
Published online Apr 24, 2020. doi: 10.5306/wjco.v11.i4.169
Narrowing the focus: Therapeutic cell surface targets for refractory triple-negative breast cancer
Narges K Tafreshi, David L Morse, Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, United States
Narges K Tafreshi, David L Morse, Department of Physics, University of South Florida, Tampa, FL 33612, United States
Narges K Tafreshi, David L Morse, Marie Catherine Lee, Division of Oncologic Sciences, University of South Florida, Tampa, FL 33612 FL, United States
Marie Catherine Lee, Comprehensive Breast Program, Moffitt Cancer Center, Tampa, FL 33612, United States
ORCID number: Narges K Tafreshi (0000-0002-2680-319X.); David L Morse (0000-0002-6006-7528); Marie Catherine Lee (0000-0002-1756-7279).
Author contributions: Tafreshi NK and Lee MC prepared initial draft of the manuscript; Morse DL contributed in the reviewing and revising the manuscript; Lee MC led the coordination in preparing the manuscript.
Conflict-of-interest statement: Authors declare no conflict of interests for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Marie Catherine Lee, FACS, MD, Surgeon, Surgical Oncologist, Comprehensive Breast Program, Moffitt Cancer Center, 12920 N. McKinley Drive, Tampa, FL 33612, United States. m.catherine.lee@moffitt.org
Received: December 30, 2019
Peer-review started: December 30, 2019
First decision: February 20, 2020
Revised: March 25, 2020
Accepted: March 28, 2020
Article in press: March 28, 2020
Published online: April 24, 2020

Abstract

Triple-negative breast cancer (TNBC) is defined as a type of breast cancer with lack of expression of estrogen receptor, progesterone receptor and human epidermal growth factor 2 protein. In comparison to other types of breast cancer, TNBC characterizes for its aggressive behavior, more prone to early recurrence and a disease with poor response to molecular target therapy. Although TNBC is identified in only 25%-30% of American breast cancer cases annually, these tumors continue to be a therapeutic challenge for clinicians for several reasons: Tumor heterogeneity, limited and toxic systemic therapy options, and often resistance to current standard therapy, characterized by progressive disease on treatment, residual tumor after cytotoxic chemotherapy, and early recurrence after complete surgical excision. Cell-surface targeted therapies have been successful for breast cancer in general, however there are currently no approved cell-surface targeted therapies specifically indicated for TNBC. Recently, several cell-surface targets have been identified as candidates for treatment of TNBC and associated targeted therapies are in development. The purpose of this work is to review the current clinical challenges posed by TNBC, the therapeutic approaches currently in use, and provide an overview of developing cell surface targeting approaches to improve outcomes for treatment resistant TNBC.

Key Words: Breast cancer, Triple negative, Biomarker, Cell surface, Targeted therapy, Chemorefractory

Core tip: Triple-negative breast cancer continues to be a challenge in breast cancer therapeutics, as these heterogeneous tumors are refractory to many effective and well-tolerated standard treatments. Even more concerning is the subpopulation of these tumors that progress even on the most aggressive therapeutic regimens. The core of this work reviews the developing approaches for treatment-refractory triple-negative breast cancer and proposes cell-surface targeting as a novel modality for targeted treatment of resistant disease.



INTRODUCTION

Over the past decade, advances in breast cancer diagnostics, classification, and treatment have significantly improved outcomes and survival, with mortality rates from breast cancer decreasing by nearly 2% per year from 2006-2015[1] However, over 40000 American women were estimated to die from breast cancer in 2018[1], despite the fact that 95% of women diagnosed with nonmetastatic breast cancer receive curative treatment in the form of surgery, systemic therapy, and or radiation[2]. In fact, breast cancer continues to be the second leading cause of cancer death in the United States, despite the steady progress in survival[1]. The fact that uptake of breast cancer treatment is very high speaks to the ongoing challenge of treatment-resistant disease.

Advances in breast-cancer specific biomarkers have the potential to overcome resistance to all aspects of breast cancer treatment, not only with regards to systemic therapy, but also for local and regional treatments like surgery and radiation. The current focus of breast cancer biomarker therapeutic research focuses on cellular mechanisms of resistance to therapy[3,4] and the development of systemic agents inhibiting cellular pathways like mTOR, CDK4/6, and AKT. This approach has high potential for systemic agents, but limited use in regional therapeutics. In contrast, cell surface biomarkers have high potential for diagnostic and therapeutic applications. The most widely recognized use of cell surface biomarker targeting targets the human epidermal growth factor 2 (Her2) cell surface marker, for which monoclonal antibody drugs such as trastuzumab have revolutionized outcomes from cancers overexpressing this particular cell surface marker. Leveraging the specificity of cell surface targeting with the addition of a cytotoxic molecule represents the next wave of cancer therapeutics, as evinced by the clinical success of the trastuzumab-emtansine antibody drug conjugate, known as ado-trastuzumab emtansine (T-DM1)[5-7]. Similar to the mechanism of T-DM1 activity, which relies upon directed delivery of a chemotherapeutic agent based on Her2neu overexpression[3], the addition of fluorescent or radioactive labelling of breast cancer specific cell surface markers may also be utilized in improving surgical visualization of tumors, or directed radiopharmaceutical use. However, cell surface markers have not been well characterized for other breast cancer subtypes or in the setting of treatment-refractory disease.

In particular, triple-negative breast cancer (TNBC) continues to pose a therapeutic challenge for women, as their outcomes are not improved by hormonal suppression or Her2neu targeted agents, and systemic therapy continues to be a backbone of standard chemotherapy agents with variable effects on short and long-term response[8]. Although TNBC is identified in only 25%-30% of American breast cancer cases annually, these tumors continue to dominate the clinical and research landscape[9]. Particularly challenging are women whose disease is refractory to standard therapy in the neoadjuvant or adjuvant setting. TNBC has a markedly poorer 5-year survival, approximately 75%, compared to 90% for hormone responsive breast cancers, with peak hazard rates for breast cancer related death at 7.5% per year 2 years after diagnosis[10]. Given the ongoing problem of treating this particular breast cancer subtype, biomarker discovery for TNBC is an ongoing area of interest among many investigators. It is important to note that TNBC is clearly an umbrella term encompassing a minority of all breast cancers, but representing a widely heterogeneous group on a molecular and biologic level, which translates to great variation in short and long-term therapeutic outcomes[8,11,12]. Recent data suggests that women who remain disease-free 5 years after treatment for TNBC have excellent disease free survival[12], so clearly not all TNBCs are the same. With this in mind, biomarker discovery for TNBCs should focus on lesions that are treatment refractory, as evinced by disease progression on chemotherapy or even residual tumor after receipt of neoadjuvant chemotherapy.

CURRENT TREATMENTS FOR RESISTANT TRIPLE-NEGATIVE BREAST CANCER

Curative, localized, TNBC is generally treated with multimodal therapy, including surgery, chemotherapy, and radiation[13]. First-line curative systemic therapy involves chemotherapy, often given in the preoperative neoadjuvant setting, as a complete pathologic response to therapy at the time of surgery confers better prognosis. Standard chemotherapy agents include Adriamycin, Cytoxan, and Taxol[13]. Additional chemotherapy agents, including carboplatin or gemcitabine, may also be considered as part of an initial therapy regimen for refractory disease; emerging data suggests that the addition of platinum-based agents may increase the rate of pathologic complete responses to chemotherapy at the time of surgical resection, and are the focus of several ongoing clinical trials[14].

Resistant disease can be identified in patients with residual disease in the breast or regional nodes at the time of surgery or may present as a recurrence either regionally or at a distant metastatic site after curative therapy. Patients with residual disease after neoadjuvant chemotherapy and surgery are now being considered for additional adjuvant systemic therapy with capecitabine, as a recent publication suggests that this is a safe adjuvant therapeutic option with improved disease free and overall survival in this population. Radiation to the breast/chest wall, with or without the nodal basins, is also generally incorporated in the treatment plan[15].

Local-regional recurrences are generally treated with surgical resection followed by radiation, and often followed with additional chemotherapy[13]. Distant metastases are generally treated systemically only, but there are no universally effective systemic therapies for long-term suppression of TNBC due to intolerance of prolonged therapy, and this population is the subject of multiple ongoing clinical trials due to the paucity of effective, tolerable therapy. In general, surgical resection or local radiation are reserved for symptomatic lesions and palliative procedures.

THE CASE FOR CELL-SURFACE TARGETING IN BREAST CANCER

The introduction of trastuzumab, which received food and drug administration (FDA) approval in September of 1998, revolutionized the approach to solid tumor treatment. Trastuzumab is a monoclonal antibody targeting Her-2, a cell-surface receptor overexpressed in approximately 30% of breast cancers, and markedly improved the disease-free survival and overall survival of women with breast cancers with overexpression of HER-2 in multiple clinical trials; this effect appears to be quite durable, with a marked improvement in outcomes even after 11 years of followup[16,17]. Trastuzumab was the first in its class of targeted cancer therapeutics; since its introduction, pertuzumab, another monoclonal inhibitor of Her-2, has been added to the standard regimen for treatment of women with Her-2 positive breast cancers in the neoadjuvant, adjuvant, and metastatic settings[7,18].

Building on the success of Her-2 targeting, T-DM1 received FDA approval in 2013 for use in treating refractory, metastatic Her2 positive breast cancers. T-DM1 is an antibody-drug conjugate, which further augments the cytotoxicity of trastuzumab by conjugation of the antibody to a tubulin inhibitor molecule, and has been demonstrated to improve outcomes for trastuzumab-resistant disease, with recent expansion to use in women with residual disease after neoadjuvant chemotherapy in combination with trastuzumab and pertuzumab[5,7,19,20].

Two cell-surface targeted antibody-drug conjugates are currently in clinical trials for pretreated and treatment refractory breast cancer. The most developed of these is Sacituzumab govitecan (IMMU-132), which is currently enrolling for an international phase III trial for treatment refractory or relapsed TNBC (ASCENT study; ClinicalTrials.gov ID NCT02574455). IMMU-132 is a conjugate of a humanized antitrophoblast cell-surface antigen 2 (Trop-2) monoclonal antibody hRS7 IgG1k to SN-38. SN-38 is an active metabolite of irinotecan, a topoisomerase I inhibitor. This is joined by a cleavable CL2A linker, which enables release of SN-38 both intracellularly as well as in the extracellular tumor microenvironment after binding to Trop-2, facilitating destruction of IMMU-132-bound cells as well as adjacent tumor cells. Trop-2 is expressed on the cell surface of 75% of TNBC patients[21].

Also, in early phase trials is ladiratuzumab vedotin (SGN-LIV1a), another antibody-drug conjugate targeting LIV-1, which is a multi-span transmembrane protein. This protein acts as a metalloproteinase as well as a zinc transporter, and is highly expressed in multiple malignancies, including metastatic estrogen-receptor positive and TNBC, as well as melanoma, prostate, and pancreatic cancer. The anti-LIV-1 antibody is linked to monomethyl auristatin E (MMAE), which disrupts microtubules[22,23]. Although LIV-1 is expressed in 65% of TNBC patients, it is expressed in normal tissue, with up to 50% 1-2 intensity staining in normal breast on IHC[22,24]. SGN-LIV1a is currently in Phase I and Phase II breast cancer trials. The Phase I trials enroll patients with metastatic/locally advanced breast cancer both in the United States (ClinicalTrials.gov identifier NCT03310957) as well as internationally (ClinicalTrials.gov identifier NCT03310957). SGN-LIV1a is also in a randomized multicenter international Phase Ib/II trial for metastatic/unresectable TNBC (Morpheus-TNBC, ClinicalTrials.gov identifier NCT3424005), as well as the adaptive randomized neoadjuvant ISPY-2 trial in the United States (ClinicalTrials.gov identifier NCT01042379), which is not limited to triple negative disease.

In addition to the targeting approaches currently being applied to breast cancer, novel targeted therapies are being developed that have shown efficacy against other cancer types that could be applied to breast cancer. For example, targeted radionuclide therapies such as Lutathera®, which has demonstrated efficacy in the treatment of mid-gut neuroendocrine tumors[25] and has been approved by the FDA, could be applied to breast cancer. Bispecific antibodies that target immune checkpoint modulating antibodies to the tumor cell-surface are another example of targeted therapies that could be applied to breast cancer[26].

TNBC CELL-SURFACE TARGETS AND TARGETED THERAPIES

Although, cell-surface targeted therapies have been successful for breast cancer in general, there are currently no approved cell-surface targeted therapies specifically indicated for TNBC. However, there are a number of cell-surface targets that have been identified as candidates for treatment of TNBC and associated targeted therapies are in development. Table 1 lists targets that have been identified as specific for TNBC and corresponding therapies are currently being tested in pre-clinical studies. Table 2 lists cell-surface targets and targeted therapies that are candidates for treatment of TNBC that have been tested in clinical trials. Of these, epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptors 1-3, GPNMB, Trop-2 receptor and LIV-1 targeted therapies have been reviewed[27-33] and only EGFR, vascular endothelial growth factor receptors 1-3 and programmed death ligand 1 have published clinical trials that specifically included patients with TNBC (Table 2).

Table 1 Pre-clinical studies targeting the cell-surface of triple negative breast cancer.
Gene, ProteinProtein type and function1TNBC expression2Normal tissue expressionDrug3Studies4Ref.
ANTXR1 (TEM8), Anthrax toxin receptor 1SPT1MP, integrin-like; Attachment, migration, progressionProtein, n = 23, 100% stromal; very low in surrounding NB. Protein, BC tissues, n = 120, NB, n = 33, increased in invasive BC6High: Gallbladder; Medium and low: Broadly expressedCAR-TXenograft regression[34-36]
ICAM1, Intercellular adhesion molecule-1SPT1MP; Binds leukocyte adhesion protein LFA-1 (integrin αL/β2)mRNA, n = 6 (cell lines), 60%; BC 25%. Protein, n = 6 (cell lines), increased expression5High: Lung, kidney. Medium: Bone marrow and immune system, endometrium. Low: Cerebral cortex, colon, bladder, testis, fallopian tubemAb: Enlimomab (murine mAb against the human ICAM1); TLipo: Lipocalin-2 siRNA payloadCAM assay; decreased xenograft angiogenesis[37,38]
MELK, Maternal embryonic leucine zipper kinasePMP, serine/threonine kinase. Cell cycle regulation, stem-cell self-renewal, apoptosis, splicing regulation, radiation resistancemRNA, n = 59, increased relative to BC, n = 284 and NB (n = 105)5High and medium: Broadly expressedIb: OTSSP167Ib + radiation decreased xenograft growth[39,40]
FZD7, Frizzled-7MPMP, Wnt protein receptor. Possibly signals polarity during morphogenesis, differentiationmRNA, n = 5, increased relative to BC, n = 145High and medium: Broadly expressedshRNA against FZD7Decreased xenograft growth[41]
MMP14, Matrix metallo-proteinase-14SPT1MP, Endopeptidase. Degrades extracellular matrixND, general increased in metastatic cancers5Medium and low: Broadly expressedHumanized Fab AbDecreased progression and metastasis of syngeneic tumors[42]
MSLN, MesothelinCell surface GPI anchor, secreted. Cell adhesionProtein, n = 109, 34%. Protein, n = 99, 67% mRNA, n = 226, increased relative to BC, n-886Broadly very low to non-expressed. 7120 organs: (1) Lung, mesothelial cells, uterus; (2) Low in heart, kidney and placentaADC: RG7787, Ab fragment/pseudomonas exotoxin A. CAR-T (TNBC not tested)Xenograft regression[43-47]
GBP1, Guanylate-binding protein 1Cell surface lipid anchor, secreted. Hydrolyzes GTP to GMP. Host protection against pathogensmRNA, n = 1512, increased relative to BC, n = 1412 and NB, n = 38875Medium: Thyroid, appendix, small intestine. Low: Brain, tonsil, lung, GI tract, kidney, fallopian tube, endometrium, skinNoneExpression controlled by EGFR. Knockdown decreased cell growth[48]
MST1R, Macrophage-stimulating protein receptor (RON)SPT1MP, tyrosine kinase receptor. MST1 ligand. Proliferation, survival, migration, differentiationProtein, n = 168, 77% expression and 45% overexpression5High: Thyroid, lung, gallbladder, ovary, placenta. Medium: Broadly expressedADC: Zt/g4- MMAE (hAb from murine mAb conjugated to MMAE)Xenograft regression[49,50]
MUC1, Mucin-1SPT1MP, extracellular or secreted. Adhesion, protective layer, progression, genotoxic stress responseProtein, n = 52, 94%5High: Lung, gallbladder, GI tract, female tissues. Medium and low: Adrenal gland, bone marrow and immune, kidney, bladder, male tissues, skinADC: mAb-MMAEPDX regression[51,52]
CDCP1,CUB domain-containing protein 1SPMP. Anchorage, migration, proliferation, differentiationProtein, n = 100, 57%5Medium and low: Broadly expressedIb: Glyco-conjugated palladium complex (Pd-Oqn)Decreased metastasis[53-56]
PIM1, Serine/threonine-protein kinase pim-1Isoform 2: Cell surface, serine/threonine kinase. Proto-oncogene. Survival, proliferation, apoptosismRNA, n = 123, increased relative to BC, n = 6475Low: Broadly expressedIb: AZD1208, PIM kinase inhibitorsStopped PDX growth; increased MYC expression; MYC-driven GEMM[57-59]
NECTIN4, Nectin-4SPT1MP. Cell adhesionmRNA, n = 1175, 61%. Protein, n = 61, 62%; NB, n = 2, 0%; ON, n = 30, 0%5Medium: Tonsil, oral mucosa, esophagus, bladder, breast, placenta, skin. Low: Pancreas, kidney, female and male tissuesADC: hAb-MMAERapid, complete, durable responses in PDXs[60]
GPR55, G-protein coupled receptor 55MPMP, LPI receptorProtein, n = 27, 82%6Broadly expressed, higher levels in bone marrow and immune system, lung, gall bladder, GI tract, bladder, female and male tissues. 772 organs, leukocyte, brain, boneshRNA against GPR55Decreased xenograft growth[61]
LRP8, Low-density lipoprotein receptor-related protein 8SPT1MP, reelin and apolipoprotein E receptormRNA, METABRIC data set, increased relative to BC5High: Testis. Low: PlacentasiRNA against LRP8; shRNA against LRP8: InducibleKnockdown in cells. Decreased tumorigenesis via Wnt signaling inhibition[62,63]
Table 2 Clinical studies targeting the TNBC cell surface.
Gene, ProteinProtein type and function1Expression in TNBC2Normal tissue expressionTargeted Drug3Clinical trialsRef.
EGFR, Epidermal growth factor receptor(1) SPT1MP, tyrosine kinase receptor; (2) EGF ligands; (3) RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCγ-PKC and STAT pathways(1) Protein, n = 316, 37%. (2) Protein, n = 930, 54%. (3) Protein, n = 17, 89%4High: Placenta. Low: Bladder, liver, skeletal muscle, skin, testis, tonsil, vaginaIb: Afatinib, Gefitinib, Lapatinib. Ab: Cetuximab, MM 151 Ab mixturePhase 2[64-71]
VGFR1-3, Vascular endothelial growth factor receptors 1-3(1) SPT1MP, tyrosine kinase receptors; (2) VEGF A,B,C,D,PGF ligands; (3) Angiogenesis, lymphangiogenesis, cell survival, migration, chemotaxis, invasion, vascular development and permeability(1) Genomic, increased copy n = 87, 62%; (2) Genomic, increased copy, n = 35, 29%6VEGF1: 220 organs- lung, placenta, liver, kidney, heart, brain. VEGF2: 208 organs, lung, cornea, broadly expressed. VEGF3: 121 organs– liver, muscle, thymus, placenta, lung, testis, ovary, prostate, heart, kidneyIb: Cediranib, Apatinib, LucitanibPhase 2[72-74]
FGFR1, Fibroblast growth factor receptor 1(1) SPT1MP, tyrosine kinase receptor; (2) FGF ligands; (3) Proliferation, migrationGenomic, increased copy, n = 76, 9%; mRNA, n = 56, 4%4High: Gallbladder, esophagus, fallopian tube, placenta. Medium and Low: Broadly expressedIb: LucitanibPhase 2[75-78]
GPNMB, Transmembrane glycoprotein NMB(1) SPT1MP; (2) Possible melanogenic enzymemRNA, n = 103, 29%5High: Skin. Medium: Cervix, uterine, gallbladder. Low: Broadly expressedADC: Glembatumumab vedotin (CDX-011)Phase 2[79]
TACSTD2, Tumor-asscociated calcium signal transducer 2 (Trop-2 receptor)(1) SPT1MP; (2) Possible growth factor receptorProtein, n = 96, 75%4Medium: Nasopharynx, bronchus, oral mucosa, esophagus, bladder, seminal vesicle, cervix, uterine, skin. Low: Multiple sitesADC: Sacituzumab govitecan (IMMU-132)Phase 2[21]
SLC39A6, Zinc transporter ZIP6 (LIV-1)(1) MPMP; (2) Possible zinc-influx transporterProtein, n = 20, 65%4High: Adrenal gland, endometrium. Medium and low: Broadly expressedADC: SGN–Ab and human Ab-MMAEPhase 1/2[22,24]
CD274, Programmed cell death 1 ligand 1 (PD-L1)(1) SPT1MP; (2) Immune tolerance, antitumor immunityProtein, n = 127, 30.7%4High: Lung, placenta. Medium: Lymph node, tonsil, spleen. Low: Appendix, colonAb: Avelumab, Atezolizumab, BMS-936559, Durvalumab, HLX20, LDP, LY3300054. CAR-T. CSR-TPhase 1, 2, 3[80-85]

As development of a targeted therapy requires a significant expense in funds and effort, there are a few major prerequisites that should be considered prior to development of a cancer targeted therapy. First, it is important to understand the intensity and breadth of expression of the target marker in the tumor tissue to be targeted, i.e., treatment resistant TNBC. Since, mRNA levels are not necessarily proportional to protein levels, a necessary step in the validation of putative targets involves the confirmation of protein levels on the surface of tumor cells in patient specimens. It is necessary to evaluate target expression in patient specimens instead of cultured cell lines, because cell lines are cultured in medium that is not representative of the tumor microenvironment and will not likely be representative of expression in patient tumors. It is notable that of the targets identified in Tables 1 and 2, only 52% (11/21) have confirmed protein expression in TNBC patient specimens, and only 24% (5/21) of these were evaluated in sample sets of n ≥ 100. Of the targets characterized with larger sample sets, protein expression was reported to be observed in 31%-77% of the samples studied. However, the intensity of expression was typically not indicated. For only 3 of these targets, elevated expression relative to surrounding normal breast tissue was reported. The ratio of expression of the target in normal tissues of concern for toxicity relative to tumor expression is another factor that can influence dose limiting toxicity and therapeutic window. Per The Human Protein Atlas, all of the targets that had confirmed protein expression in patient TNBC specimens, had medium or high expression in tissues of concern for toxicity or were broadly expressed in normal tissues. Exceptions were EGFR which only has low expression in the liver and mesothelin which only has low expression in the bronchus, nasopharynx and oral mucosa. Finally, it is important that the target expression be representative of the untreatable fraction of TNBC, i.e., the patients with resistant disease. None of the target identification studies included TNBC specimens known to be resistant to standard therapy.

CONCLUSION

Novel effective therapies are needed for the treatment of chemotherapy resistant TNBC which has an extremely poor prognosis. Cell-surface targeted therapies have demonstrated efficacy in the treatment of breast cancer in general, e.g., the Her2 inhibiting antibody Trastuzumab, or the antibody-drug conjugate T-DM1. However, there are no targeted therapies that are specific for the effective treatment of resistant TNBC. Although some TNBC targets have been identified, few have been well characterized in terms of intensity and breadth of expression in TNBC patient specimens, nor in terms of expression in normal tissues of concern for toxicity. The ratio of expression in tumor versus normal tissues is a key factor in the therapeutic window observed for a corresponding targeted therapy. Some protection of normal tissues may be observed due to the relatively low permeability of vasculature in most normal tissues relative to tumor vasculature. However, this is not the case for some key clearance organs, i.e., kidney and liver, which also have permeable vasculature. Systematic studies to discover cell-surface therapeutic targets for resistant TNBC are greatly needed. Once validated, novel and effective targeted therapies may be developed for resistant TNBC tumors and metastases.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Oncology

Country/Territory of origin: United States

Peer-review report´s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Angelucci A, Cidon EU, Scaggiante B S-Editor: Dou Y L-Editor: A E-Editor: Zhang YL

References
1.  Balko JM, Giltnane JM, Wang K, Schwarz LJ, Young CD, Cook RS, Owens P, Sanders ME, Kuba MG, Sánchez V, Kurupi R, Moore PD, Pinto JA, Doimi FD, Gómez H, Horiuchi D, Goga A, Lehmann BD, Bauer JA, Pietenpol JA, Ross JS, Palmer GA, Yelensky R, Cronin M, Miller VA, Stephens PJ, Arteaga CL. Molecular profiling of the residual disease of triple-negative breast cancers after neoadjuvant chemotherapy identifies actionable therapeutic targets. Cancer Discov. 2014;4:232-245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 365]  [Article Influence: 33.2]  [Reference Citation Analysis (0)]
2.  Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R, Jemal A. Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 2016;66:271-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3134]  [Cited by in F6Publishing: 3380]  [Article Influence: 422.5]  [Reference Citation Analysis (0)]
3.  Tang Y, Wang Y, Kiani MF, Wang B. Classification, Treatment Strategy, and Associated Drug Resistance in Breast Cancer. Clin Breast Cancer. 2016;16:335-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 160]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
4.  Brufsky AM. Long-term management of patients with hormone receptor-positive metastatic breast cancer: Concepts for sequential and combination endocrine-based therapies. Cancer Treat Rev. 2017;59:22-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
5.  von Minckwitz G, Huang CS, Mano MS, Loibl S, Mamounas EP, Untch M, Wolmark N, Rastogi P, Schneeweiss A, Redondo A, Fischer HH, Jacot W, Conlin AK, Arce-Salinas C, Wapnir IL, Jackisch C, DiGiovanna MP, Fasching PA, Crown JP, Wülfing P, Shao Z, Rota Caremoli E, Wu H, Lam LH, Tesarowski D, Smitt M, Douthwaite H, Singel SM, Geyer CE; KATHERINE Investigators. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N Engl J Med. 2019;380:617-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1122]  [Cited by in F6Publishing: 1348]  [Article Influence: 269.6]  [Reference Citation Analysis (0)]
6.  Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram M, Oh DY, Diéras V, Guardino E, Fang L, Lu MW, Olsen S, Blackwell K; EMILIA Study Group. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783-1791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2411]  [Cited by in F6Publishing: 2459]  [Article Influence: 204.9]  [Reference Citation Analysis (0)]
7.  Hurvitz SA, Martin M, Symmans WF, Jung KH, Huang CS, Thompson AM, Harbeck N, Valero V, Stroyakovskiy D, Wildiers H, Campone M, Boileau JF, Beckmann MW, Afenjar K, Fresco R, Helms HJ, Xu J, Lin YG, Sparano J, Slamon D. Neoadjuvant trastuzumab, pertuzumab, and chemotherapy versus trastuzumab emtansine plus pertuzumab in patients with HER2-positive breast cancer (KRISTINE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol. 2018;19:115-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 276]  [Article Influence: 39.4]  [Reference Citation Analysis (0)]
8.  Gutzmer R, Rivoltini L, Levchenko E, Testori A, Utikal J, Ascierto PA, Demidov L, Grob JJ, Ridolfi R, Schadendorf D, Queirolo P, Santoro A, Loquai C, Dreno B, Hauschild A, Schultz E, Lesimple TP, Vanhoutte N, Salaun B, Gillet M, Jarnjak S, De Sousa Alves PM, Louahed J, Brichard VG, Lehmann FF. Safety and immunogenicity of the PRAME cancer immunotherapeutic in metastatic melanoma: results of a phase I dose escalation study. ESMO Open. 2016;1:e000068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 50]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
9.  Boyle P. Triple-negative breast cancer: epidemiological considerations and recommendations. Ann Oncol. 2012;23 Suppl 6:vi7-v12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 371]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
10.  Gierach GL, Burke A, Anderson WF. Epidemiology of triple negative breast cancers. Breast Dis. 2010;32:5-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 40]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
11.  Prat A, Adamo B, Cheang MC, Anders CK, Carey LA, Perou CM. Molecular characterization of basal-like and non-basal-like triple-negative breast cancer. Oncologist. 2013;18:123-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 361]  [Cited by in F6Publishing: 389]  [Article Influence: 35.4]  [Reference Citation Analysis (0)]
12.  Reddy SM, Barcenas CH, Sinha AK, Hsu L, Moulder SL, Tripathy D, Hortobagyi GN, Valero V. Long-term survival outcomes of triple-receptor negative breast cancer survivors who are disease free at 5 years and relationship with low hormone receptor positivity. Br J Cancer. 2018;118:17-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 57]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
13.  NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®): Breast Cancer.  Plymouth Meeting, PA: National Comprehensive Cancer Network, 2019.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Walsh EM, Shalaby A, O'Loughlin M, Keane N, Webber MJ, Kerin MJ, Keane MM, Glynn SA, Callagy GM. Outcome for triple negative breast cancer in a retrospective cohort with an emphasis on response to platinum-based neoadjuvant therapy. Breast Cancer Res Treat. 2019;174:1-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
15.  Masuda N, Lee SJ, Ohtani S, Im YH, Lee ES, Yokota I, Kuroi K, Im SA, Park BW, Kim SB, Yanagita Y, Ohno S, Takao S, Aogi K, Iwata H, Jeong J, Kim A, Park KH, Sasano H, Ohashi Y, Toi M. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med. 2017;376:2147-2159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 893]  [Cited by in F6Publishing: 1011]  [Article Influence: 144.4]  [Reference Citation Analysis (0)]
16.  Smith I, Procter M, Gelber RD, Guillaume S, Feyereislova A, Dowsett M, Goldhirsch A, Untch M, Mariani G, Baselga J, Kaufmann M, Cameron D, Bell R, Bergh J, Coleman R, Wardley A, Harbeck N, Lopez RI, Mallmann P, Gelmon K, Wilcken N, Wist E, Sánchez Rovira P, Piccart-Gebhart MJ; HERA study team. 2-year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: a randomised controlled trial. Lancet. 2007;369:29-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1121]  [Cited by in F6Publishing: 1043]  [Article Influence: 61.4]  [Reference Citation Analysis (0)]
17.  Cameron D, Piccart-Gebhart MJ, Gelber RD, Procter M, Goldhirsch A, de Azambuja E, Castro G, Untch M, Smith I, Gianni L, Baselga J, Al-Sakaff N, Lauer S, McFadden E, Leyland-Jones B, Bell R, Dowsett M, Jackisch C; Herceptin Adjuvant (HERA) Trial Study Team. 11 years' follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin Adjuvant (HERA) trial. Lancet. 2017;389:1195-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 572]  [Cited by in F6Publishing: 626]  [Article Influence: 89.4]  [Reference Citation Analysis (0)]
18.  von Minckwitz G, Procter M, de Azambuja E, Zardavas D, Benyunes M, Viale G, Suter T, Arahmani A, Rouchet N, Clark E, Knott A, Lang I, Levy C, Yardley DA, Bines J, Gelber RD, Piccart M, Baselga J; APHINITY Steering Committee and Investigators. Adjuvant Pertuzumab and Trastuzumab in Early HER2-Positive Breast Cancer. N Engl J Med. 2017;377:122-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 918]  [Cited by in F6Publishing: 867]  [Article Influence: 123.9]  [Reference Citation Analysis (0)]
19.  Montemurro F, Ellis P, Anton A, Wuerstlein R, Delaloge S, Bonneterre J, Quenel-Tueux N, Linn SC, Irahara N, Donica M, Lindegger N, Barrios CH. Safety of trastuzumab emtansine (T-DM1) in patients with HER2-positive advanced breast cancer: Primary results from the KAMILLA study cohort 1. Eur J Cancer. 2019;109:92-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 62]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
20.  Burris HA, Rugo HS, Vukelja SJ, Vogel CL, Borson RA, Limentani S, Tan-Chiu E, Krop IE, Michaelson RA, Girish S, Amler L, Zheng M, Chu YW, Klencke B, O'Shaughnessy JA. Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy. J Clin Oncol. 2011;29:398-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 496]  [Cited by in F6Publishing: 510]  [Article Influence: 36.4]  [Reference Citation Analysis (0)]
21.  Zhao W, Kuai X, Zhou X, Jia L, Wang J, Yang X, Tian Z, Wang X, Lv Q, Wang B, Zhao Y, Huang W. Trop2 is a potential biomarker for the promotion of EMT in human breast cancer. Oncol Rep. 2018;40:759-766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 23]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
22.  Sussman D, Smith LM, Anderson ME, Duniho S, Hunter JH, Kostner H, Miyamoto JB, Nesterova A, Westendorf L, Van Epps HA, Whiting N, Benjamin DR. SGN-LIV1A: a novel antibody-drug conjugate targeting LIV-1 for the treatment of metastatic breast cancer. Mol Cancer Ther. 2014;13:2991-3000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 86]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
23.  Kostic A, Anderson M, Duniho S, Miyamoto J, Nesterova A, Sussman D. SGN-LIV1A, an antibody-drug conjugate (ADC), in patients with LIV-1–positive breast cancer. J Clin Oncol. 2014;32 suppl 15:TPS1143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
24.  Doronina SO, Toki BE, Torgov MY, Mendelsohn BA, Cerveny CG, Chace DF, DeBlanc RL, Gearing RP, Bovee TD, Siegall CB, Francisco JA, Wahl AF, Meyer DL, Senter PD. Development of potent monoclonal antibody auristatin conjugates for cancer therapy. Nat Biotechnol. 2003;21:778-784.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 843]  [Cited by in F6Publishing: 799]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
25.  Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen B, Mittra E, Kunz PL, Kulke MH, Jacene H, Bushnell D, O'Dorisio TM, Baum RP, Kulkarni HR, Caplin M, Lebtahi R, Hobday T, Delpassand E, Van Cutsem E, Benson A, Srirajaskanthan R, Pavel M, Mora J, Berlin J, Grande E, Reed N, Seregni E, Öberg K, Lopera Sierra M, Santoro P, Thevenet T, Erion JL, Ruszniewski P, Kwekkeboom D, Krenning E; NETTER-1 Trial Investigators. Phase 3 Trial of 177Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med. 2017;376:125-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1702]  [Cited by in F6Publishing: 1858]  [Article Influence: 265.4]  [Reference Citation Analysis (0)]
26.  Yu L, Wang J. T cell-redirecting bispecific antibodies in cancer immunotherapy: recent advances. J Cancer Res Clin Oncol. 2019;145:941-956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
27.  Shao F, Sun H, Deng CX. Potential therapeutic targets of triple-negative breast cancer based on its intrinsic subtype. Oncotarget. 2017;8:73329-73344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
28.  Zhang JF, Liu J, Wang Y, Zhang B. Novel therapeutic strategies for patients with triple-negative breast cancer. Onco Targets Ther. 2016;9:6519-6528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
29.  Rose AAN, Biondini M, Curiel R, Siegel PM. Targeting GPNMB with glembatumumab vedotin: Current developments and future opportunities for the treatment of cancer. Pharmacol Ther. 2017;179:127-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
30.  Shvartsur A, Bonavida B. Trop2 and its overexpression in cancers: regulation and clinical/therapeutic implications. Genes Cancer. 2015;6:84-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 181]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
31.  Zaman S, Jadid H, Denson AC, Gray JE. Targeting Trop-2 in solid tumors: future prospects. Onco Targets Ther. 2019;12:1781-1790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 80]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
32.  Starodub AN, Ocean AJ, Shah MA, Guarino MJ, Picozzi VJ, Vahdat LT, Thomas SS, Govindan SV, Maliakal PP, Wegener WA, Hamburger SA, Sharkey RM, Goldenberg DM. First-in-Human Trial of a Novel Anti-Trop-2 Antibody-SN-38 Conjugate, Sacituzumab Govitecan, for the Treatment of Diverse Metastatic Solid Tumors. Clin Cancer Res. 2015;21:3870-3878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 201]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
33.  Taylor KM. A distinct role in breast cancer for two LIV-1 family zinc transporters. Biochem Soc Trans. 2008;36:1247-1251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
34.  Gutwein LG, Al-Quran SZ, Fernando S, Fletcher BS, Copeland EM, Grobmyer SR. Tumor endothelial marker 8 expression in triple-negative breast cancer. Anticancer Res. 2011;31:3417-3422.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Byrd TT, Fousek K, Pignata A, Szot C, Samaha H, Seaman S, Dobrolecki L, Salsman VS, Oo HZ, Bielamowicz K, Landi D, Rainusso N, Hicks J, Powell S, Baker ML, Wels WS, Koch J, Sorensen PH, Deneen B, Ellis MJ, Lewis MT, Hegde M, Fletcher BS, St Croix B, Ahmed N. TEM8/ANTXR1-Specific CAR T Cells as a Targeted Therapy for Triple-Negative Breast Cancer. Cancer Res. 2018;78:489-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 110]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
36.  Davies G, Rmali KA, Watkins G, Mansel RE, Mason MD, Jiang WG. Elevated levels of tumour endothelial marker-8 in human breast cancer and its clinical significance. Int J Oncol. 2006;29:1311-1317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
37.  Guo P, Huang J, Wang L, Jia D, Yang J, Dillon DA, Zurakowski D, Mao H, Moses MA, Auguste DT. ICAM-1 as a molecular target for triple negative breast cancer. Proc Natl Acad Sci USA. 2014;111:14710-14715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 133]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
38.  Guo P, Yang J, Jia D, Moses MA, Auguste DT. ICAM-1-Targeted, Lcn2 siRNA-Encapsulating Liposomes are Potent Anti-angiogenic Agents for Triple Negative Breast Cancer. Theranostics. 2016;6:1-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 91]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
39.  Speers C, Zhao SG, Kothari V, Santola A, Liu M, Wilder-Romans K, Evans J, Batra N, Bartelink H, Hayes DF, Lawrence TS, Brown PH, Pierce LJ, Feng FY. Maternal Embryonic Leucine Zipper Kinase (MELK) as a Novel Mediator and Biomarker of Radioresistance in Human Breast Cancer. Clin Cancer Res. 2016;22:5864-5875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 78]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
40.  Moreno CS. MELK kinase holds promise as a new radiosensitizing target and biomarker in triple-negative breast cancer. J Thorac Dis. 2016;8:E1367-E1368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
41.  Yang L, Wu X, Wang Y, Zhang K, Wu J, Yuan YC, Deng X, Chen L, Kim CC, Lau S, Somlo G, Yen Y. FZD7 has a critical role in cell proliferation in triple negative breast cancer. Oncogene. 2011;30:4437-4446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 200]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
42.  Ling B, Watt K, Banerjee S, Newsted D, Truesdell P, Adams J, Sidhu SS, Craig AWB. A novel immunotherapy targeting MMP-14 limits hypoxia, immune suppression and metastasis in triple-negative breast cancer models. Oncotarget. 2017;8:58372-58385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
43.  Parinyanitikul N, Blumenschein GR, Wu Y, Lei X, Chavez-Macgregor M, Smart M, Gonzalez-Angulo AM. Mesothelin expression and survival outcomes in triple receptor negative breast cancer. Clin Breast Cancer. 2013;13:378-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
44.  Tchou J, Wang LC, Selven B, Zhang H, Conejo-Garcia J, Borghaei H, Kalos M, Vondeheide RH, Albelda SM, June CH, Zhang PJ. Mesothelin, a novel immunotherapy target for triple negative breast cancer. Breast Cancer Res Treat. 2012;133:799-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 113]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
45.  Tozbikian G, Brogi E, Kadota K, Catalano J, Akram M, Patil S, Ho AY, Reis-Filho JS, Weigelt B, Norton L, Adusumilli PS, Wen HY. Mesothelin expression in triple negative breast carcinomas correlates significantly with basal-like phenotype, distant metastases and decreased survival. PLoS One. 2014;9:e114900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 73]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
46.  Alewine C, Xiang L, Yamori T, Niederfellner G, Bosslet K, Pastan I. Efficacy of RG7787, a next-generation mesothelin-targeted immunotoxin, against triple-negative breast and gastric cancers. Mol Cancer Ther. 2014;13:2653-2661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
47.  Morello A, Sadelain M, Adusumilli PS. Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors. Cancer Discov. 2016;6:133-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 324]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
48.  Quintero M, Adamoski D, Reis LMD, Ascenção CFR, Oliveira KRS, Gonçalves KA, Dias MM, Carazzolle MF, Dias SMG. Guanylate-binding protein-1 is a potential new therapeutic target for triple-negative breast cancer. BMC Cancer. 2017;17:727.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
49.  Suthe SR, Yao HP, Weng TH, Hu CY, Feng L, Wu ZG, Wang MH. RON Receptor Tyrosine Kinase as a Therapeutic Target for Eradication of Triple-Negative Breast Cancer: Efficacy of Anti-RON ADC Zt/g4-MMAE. Mol Cancer Ther. 2018;17:2654-2664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
50.  Yao HP, Feng L, Suthe SR, Chen LH, Weng TH, Hu CY, Jun ES, Wu ZG, Wang WL, Kim SC, Tong XM, Wang MH. Therapeutic efficacy, pharmacokinetic profiles, and toxicological activities of humanized antibody-drug conjugate Zt/g4-MMAE targeting RON receptor tyrosine kinase for cancer therapy. J Immunother Cancer. 2019;7:75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
51.  Siroy A, Abdul-Karim FW, Miedler J, Fong N, Fu P, Gilmore H, Baar J. MUC1 is expressed at high frequency in early-stage basal-like triple-negative breast cancer. Hum Pathol. 2013;44:2159-2166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
52.  Panchamoorthy G, Jin C, Raina D, Bharti A, Yamamoto M, Adeebge D, Zhao Q, Bronson R, Jiang S, Li L, Suzuki Y, Tagde A, Ghoroghchian PP, Wong KK, Kharbanda S, Kufe D. Targeting the human MUC1-C oncoprotein with an antibody-drug conjugate. JCI Insight. 2018;3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 44]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
53.  Turdo F, Bianchi F, Gasparini P, Sandri M, Sasso M, De Cecco L, Forte L, Casalini P, Aiello P, Sfondrini L, Agresti R, Carcangiu ML, Plantamura I, Sozzi G, Tagliabue E, Campiglio M. CDCP1 is a novel marker of the most aggressive human triple-negative breast cancers. Oncotarget. 2016;7:69649-69665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
54.  Nakashima K, Uekita T, Yano S, Kikuchi JI, Nakanishi R, Sakamoto N, Fukumoto K, Nomoto A, Kawamoto K, Shibahara T, Yamaguchi H, Sakai R. Novel small molecule inhibiting CDCP1-PKCδ pathway reduces tumor metastasis and proliferation. Cancer Sci. 2017;108:1049-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
55.  Wright HJ, Police AM, Razorenova OV. Targeting CDCP1 dimerization in triple-negative breast cancer. Cell Cycle. 2016;15:2385-2386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
56.  Wright HJ, Hou J, Xu B, Cortez M, Potma EO, Tromberg BJ, Razorenova OV. CDCP1 drives triple-negative breast cancer metastasis through reduction of lipid-droplet abundance and stimulation of fatty acid oxidation. Proc Natl Acad Sci USA. 2017;114:E6556-E6565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 112]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
57.  Brasó-Maristany F, Filosto S, Catchpole S, Marlow R, Quist J, Francesch-Domenech E, Plumb DA, Zakka L, Gazinska P, Liccardi G, Meier P, Gris-Oliver A, Cheang MCU, Perdrix-Rosell A, Shafat M, Noël E, Patel N, McEachern K, Scaltriti M, Castel P, Noor F, Buus R, Mathew S, Watkins J, Serra V, Marra P, Grigoriadis A, Tutt AN. Erratum: PIM1 kinase regulates cell death, tumor growth and chemotherapy response in triple-negative breast cancer. Nat Med. 2017;23:788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
58.  Zhao W, Qiu R, Li P, Yang J. PIM1: a promising target in patients with triple-negative breast cancer. Med Oncol. 2017;34:142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
59.  Horiuchi D, Camarda R, Zhou AY, Yau C, Momcilovic O, Balakrishnan S, Corella AN, Eyob H, Kessenbrock K, Lawson DA, Marsh LA, Anderton BN, Rohrberg J, Kunder R, Bazarov AV, Yaswen P, McManus MT, Rugo HS, Werb Z, Goga A. PIM1 kinase inhibition as a targeted therapy against triple-negative breast tumors with elevated MYC expression. Nat Med. 2016;22:1321-1329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 117]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
60.  M-Rabet M, Cabaud O, Josselin E, Finetti P, Castellano R, Farina A, Agavnian-Couquiaud E, Saviane G, Collette Y, Viens P, Gonçalves A, Ginestier C, Charafe-Jauffret E, Birnbaum D, Olive D, Bertucci F, Lopez M. Nectin-4: a new prognostic biomarker for efficient therapeutic targeting of primary and metastatic triple-negative breast cancer. Ann Oncol. 2017;28:769-776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
61.  Andradas C, Blasco-Benito S, Castillo-Lluva S, Dillenburg-Pilla P, Diez-Alarcia R, Juanes-García A, García-Taboada E, Hernando-Llorente R, Soriano J, Hamann S, Wenners A, Alkatout I, Klapper W, Rocken C, Bauer M, Arnold N, Quintanilla M, Megías D, Vicente-Manzanares M, Urigüen L, Gutkind JS, Guzmán M, Pérez-Gómez E, Sánchez C. Activation of the orphan receptor GPR55 by lysophosphatidylinositol promotes metastasis in triple-negative breast cancer. Oncotarget. 2016;7:47565-47575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
62.  Lin CC, Lo MC, Moody R, Jiang H, Harouaka R, Stevers N, Tinsley S, Gasparyan M, Wicha M, Sun D. Targeting LRP8 inhibits breast cancer stem cells in triple-negative breast cancer. Cancer Lett. 2018;438:165-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
63.  Maire V, Mahmood F, Rigaill G, Ye M, Brisson A, Némati F, Gentien D, Tucker GC, Roman-Roman S, Dubois T. LRP8 is overexpressed in estrogen-negative breast cancers and a potential target for these tumors. Cancer Med. 2019;8:325-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
64.  Baselga J, Albanell J, Ruiz A, Lluch A, Gascón P, Guillém V, González S, Sauleda S, Marimón I, Tabernero JM, Koehler MT, Rojo F. Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J Clin Oncol. 2005;23:5323-5333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 265]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
65.  Baselga J, Gómez P, Greil R, Braga S, Climent MA, Wardley AM, Kaufman B, Stemmer SM, Pêgo A, Chan A, Goeminne JC, Graas MP, Kennedy MJ, Ciruelos Gil EM, Schneeweiss A, Zubel A, Groos J, Melezínková H, Awada A. Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J Clin Oncol. 2013;31:2586-2592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 273]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
66.  Bernsdorf M, Ingvar C, Jörgensen L, Tuxen MK, Jakobsen EH, Saetersdal A, Kimper-Karl ML, Kroman N, Balslev E, Ejlertsen B. Effect of adding gefitinib to neoadjuvant chemotherapy in estrogen receptor negative early breast cancer in a randomized phase II trial. Breast Cancer Res Treat. 2011;126:463-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 49]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
67.  Carey LA, Rugo HS, Marcom PK, Mayer EL, Esteva FJ, Ma CX, Liu MC, Storniolo AM, Rimawi MF, Forero-Torres A, Wolff AC, Hobday TJ, Ivanova A, Chiu WK, Ferraro M, Burrows E, Bernard PS, Hoadley KA, Perou CM, Winer EP. TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol. 2012;30:2615-2623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 367]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
68.  Changavi AA, Shashikala A, Ramji AS. Epidermal Growth Factor Receptor Expression in Triple Negative and Nontriple Negative Breast Carcinomas. J Lab Physicians. 2015;7:79-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 50]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
69.  Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 2004;10:5367-5374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1840]  [Cited by in F6Publishing: 1848]  [Article Influence: 97.3]  [Reference Citation Analysis (0)]
70.  Rakha EA, El-Sayed ME, Green AR, Lee AH, Robertson JF, Ellis IO. Prognostic markers in triple-negative breast cancer. Cancer. 2007;109:25-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 874]  [Cited by in F6Publishing: 894]  [Article Influence: 52.6]  [Reference Citation Analysis (0)]
71.  Rakha EA, Elsheikh SE, Aleskandarany MA, Habashi HO, Green AR, Powe DG, El-Sayed ME, Benhasouna A, Brunet JS, Akslen LA, Evans AJ, Blamey R, Reis-Filho JS, Foulkes WD, Ellis IO. Triple-negative breast cancer: distinguishing between basal and nonbasal subtypes. Clin Cancer Res. 2009;15:2302-2310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 353]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
72.  Andre F, Job B, Dessen P, Tordai A, Michiels S, Liedtke C, Richon C, Yan K, Wang B, Vassal G, Delaloge S, Hortobagyi GN, Symmans WF, Lazar V, Pusztai L. Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomic hybridization array. Clin Cancer Res. 2009;15:441-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 244]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
73.  Linderholm BK, Hellborg H, Johansson U, Elmberger G, Skoog L, Lehtiö J, Lewensohn R. Significantly higher levels of vascular endothelial growth factor (VEGF) and shorter survival times for patients with primary operable triple-negative breast cancer. Ann Oncol. 2009;20:1639-1646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 235]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
74.  Liu JF, Tolaney SM, Birrer M, Fleming GF, Buss MK, Dahlberg SE, Lee H, Whalen C, Tyburski K, Winer E, Ivy P, Matulonis UA. A Phase 1 trial of the poly(ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti-angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. Eur J Cancer. 2013;49:2972-2978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 132]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
75.  Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8940]  [Cited by in F6Publishing: 8662]  [Article Influence: 721.8]  [Reference Citation Analysis (0)]
76.  Turner N, Lambros MB, Horlings HM, Pearson A, Sharpe R, Natrajan R, Geyer FC, van Kouwenhove M, Kreike B, Mackay A, Ashworth A, van de Vijver MJ, Reis-Filho JS. Integrative molecular profiling of triple negative breast cancers identifies amplicon drivers and potential therapeutic targets. Oncogene. 2010;29:2013-2023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 317]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
77.  Soria JC, DeBraud F, Bahleda R, Adamo B, Andre F, Dienstmann R, Delmonte A, Cereda R, Isaacson J, Litten J, Allen A, Dubois F, Saba C, Robert R, D'Incalci M, Zucchetti M, Camboni MG, Tabernero J. Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, and pharmacodynamics of lucitanib in advanced solid tumors. Ann Oncol. 2014;25:2244-2251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 141]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
78.  Abu-Khalaf MM, Mayer IA, Tankersley C, Moy J, Allen AR, Vogel CL, Holmes FA, Nanda R, Miller K, Patel R, Pusztai L, Arteaga CL. A phase 2, randomized, open-label study of lucitanib in patients with FGF aberrant metastatic breast cancer. J Clin Oncol. 2015;33 suppl 15:TPS628.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Rose AA, Grosset AA, Dong Z, Russo C, Macdonald PA, Bertos NR, St-Pierre Y, Simantov R, Hallett M, Park M, Gaboury L, Siegel PM. Glycoprotein nonmetastatic B is an independent prognostic indicator of recurrence and a novel therapeutic target in breast cancer. Clin Cancer Res. 2010;16:2147-2156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 136]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
80.  Cortés J, André F, Gonçalves A, Kümmel S, Martín M, Schmid P, Schuetz F, Swain SM, Easton V, Pollex E, Deurloo R, Dent R. IMpassion132 Phase III trial: atezolizumab and chemotherapy in early relapsing metastatic triple-negative breast cancer. Future Oncol. 2019;15:1951-1961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
81.  Hwang SY, Park S, Kwon Y. Recent therapeutic trends and promising targets in triple negative breast cancer. Pharmacol Ther. 2019;199:30-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 132]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
82.  Jia H, Truica CI, Wang B, Wang Y, Ren X, Harvey HA, Song J, Yang JM. Immunotherapy for triple-negative breast cancer: Existing challenges and exciting prospects. Drug Resist Updat. 2017;32:1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 109]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
83.  Muenst S, Schaerli AR, Gao F, Däster S, Trella E, Droeser RA, Muraro MG, Zajac P, Zanetti R, Gillanders WE, Weber WP, Soysal SD. Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat. 2014;146:15-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 347]  [Cited by in F6Publishing: 423]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]
84.  Nanda R, Chow LQ, Dees EC, Berger R, Gupta S, Geva R, Pusztai L, Pathiraja K, Aktan G, Cheng JD, Karantza V, Buisseret L. Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J Clin Oncol. 2016;34:2460-2467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 866]  [Cited by in F6Publishing: 1032]  [Article Influence: 129.0]  [Reference Citation Analysis (0)]
85.  Vikas P, Borcherding N, Zhang W. The clinical promise of immunotherapy in triple-negative breast cancer. Cancer Manag Res. 2018;10:6823-6833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 92]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]