1
|
Li G, Du R, Wang D, Zhang X, Wang L, Pu S, Li X, Wang S, Zhang J, Liu B, Gao Y, Zhao H. Improved Efficacy of Triple-Negative Breast Cancer Immunotherapy via Hydrogel-Based Co-Delivery of CAR-T Cells and Mitophagy Agonist. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2409835. [PMID: 39840546 DOI: 10.1002/advs.202409835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/23/2024] [Indexed: 01/23/2025]
Abstract
Leaky and structurally abnormal blood vessels and increased pressure in the tumor interstitium reduce the infiltration of CAR-T cells in solid tumors, including triple-negative breast cancer (TNBC). Furthermore, high burden of tumor cells may cause reduction of infiltrating CAR-T cells and their functional exhaustion. In this study, various effector-to-target (E:T) ratio experiments are established to model the treatment using CAR-T cells in leukemia (high E:T ratio) and solid tumor (low E:T ratio). It is found that the antitumor immune response is decreased in solid tumors with low E:T ratio. Furthermore, single cell sequencing is performed to investigate the functional exhaustion at a low ratio. It is revealed that the inhibition of mitophagy-mediated mitochondrial dysfunction diminished the antitumor efficacy of CAR-T-cell therapy. The mitophagy agonist BC1618 is screened via AI-deep learning and cytokine detection, in vivo and in vitro studies revealed that BC1618 significantly strengthened the antitumor response of CAR-T cells via improving mitophagy. Here, injection hydrogels are engineered for the controlled co-delivery of CAR-T cells and BC1618 that improves the treatment of TNBC. Local delivery of hydrogels creates an inflammatory and mitophagy-enhanced microenvironment at the tumor site, which stimulates the CAR-T cells proliferation, provides antitumor ability persistently, and improves the effect of treatment.
Collapse
Affiliation(s)
- Guodong Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Ruoxin Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Donghui Wang
- Department of Thyroid, Breast, and Vascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, P. R. China
| | - Xiangmei Zhang
- Hebei Provincial Cancer Institute, Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, P. R. China
| | - Lizhuo Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Shuangpeng Pu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Xiaoju Li
- Bioinformatics Center of AMMS, Beijing, 100850, P. R. China
| | - Shuning Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, P. R. China
- Research Institution, Xijing hospital, The Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Juliang Zhang
- Department of Thyroid, Breast, and Vascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, P. R. China
| | - Beichen Liu
- Department of Hematology, Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, P. R. China
| | - Yuan Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, P. R. China
| |
Collapse
|
2
|
Yüceer RO, Aydın S, Gelir I, Koc T, Tuncer E, Ucar M. Exploring the Prognostic Role of Trop-2, CD47, and CD163 Expression Levels on Survival Outcomes in Patients with Triple-Negative Breast Cancer. Diagnostics (Basel) 2025; 15:232. [PMID: 39857116 PMCID: PMC11764053 DOI: 10.3390/diagnostics15020232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Background: This study evaluated the prognostic impact of Trop-2, CD47, and CD163 expression on clinical outcomes in triple-negative breast cancer (TNBC) and investigated their interactions with tumor progression. Methods: A retrospective cohort of 92 patients with TNBC was analyzed. The expression scores for Trop-2, CD47, and CD163 were categorized as negative/low (0-3 points) or high (4-6 points). The primary endpoint was overall survival (OS). Results: The median age of the cohort was 50 years old. High Trop-2 expression was observed in 55.4% of the patients and was significantly associated with advanced disease stage (p < 0.001). High CD47 expression (44.6%) was correlated with advanced stage (p = 0.044), whereas high CD163 expression (45.7%) was associated with advanced stage (p = 0.021), absence of comorbidities (p = 0.022), and lower pT stage (p = 0.023). Moderate positive correlations were found between Trop-2 and CD47 (p = 0.022), Trop-2 and CD163 (p = 0.037), and CD47 and CD163 (p < 0.001), respectively. Kaplan-Meier survival analysis revealed that patients with low Trop-2 expression exhibited significantly prolonged OS (p = 0.021) and progression-free survival (PFS) (p = 0.026) compared to those with high Trop-2 expression. Univariate and multivariate analyses revealed significant associations between OS and PFS for Trop-2, lymphovascular invasion, and BRCA status. Conclusions: Trop-2 expression is a significant prognostic factor for TNBC and is correlated with worse outcomes. Although CD47 and CD163 showed trends for poorer prognosis, their significance was not confirmed. These findings offer promising prospects for future studies on combined antibody-drug conjugates (ADCs), as they may present opportunities to address multiple resistance mechanisms in the management of TNBC and enhance clinical outcomes.
Collapse
Affiliation(s)
- Ramazan Oguz Yüceer
- Department of Pathology, Sivas Cumhuriyet University School of Medicine, Sivas 58140, Turkey; (S.A.); (T.K.); (E.T.)
| | - Sedanur Aydın
- Department of Pathology, Sivas Cumhuriyet University School of Medicine, Sivas 58140, Turkey; (S.A.); (T.K.); (E.T.)
| | - Iclal Gelir
- Sivas Cumhuriyet University School of Medicine, Sivas 58140, Turkey;
| | - Tulay Koc
- Department of Pathology, Sivas Cumhuriyet University School of Medicine, Sivas 58140, Turkey; (S.A.); (T.K.); (E.T.)
| | - Ersin Tuncer
- Department of Pathology, Sivas Cumhuriyet University School of Medicine, Sivas 58140, Turkey; (S.A.); (T.K.); (E.T.)
| | - Mahmut Ucar
- Department of Medical Oncology, Sivas Cumhuriyet University School of Medicine, Sivas 58140, Turkey;
| |
Collapse
|
3
|
Vozgirdaite D, Allard-Vannier E, Velge-Roussel F, Douez E, Jolivet L, Boursin F, Chourpa I, Aubrey N, Hervé-Aubert K. Metformin-encapsulating immunoliposomes conjugated with anti-TROP 2 antibody fragments for the active targeting of triple-negative breast cancer. NANOSCALE 2025. [PMID: 39775761 DOI: 10.1039/d4nr03224a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Trophoblast cell-surface antigen 2 (TROP 2) has re-emerged as a promising biomarker in triple-negative breast cancer (TNBC), with high overexpression in many TNBC cases. However, despite its potential and approval as an antibody-drug-conjugate for TNBC treatment, TROP 2-targeted delivery systems are currently underexplored. Therefore, this study was aimed at exploiting the potential of TROP 2 targeting by encapsulating metformin (Met), an antidiabetic drug associated with tumor growth inhibitory properties, inside liposomes decorated with TROP 2-targeting single-chain variable fragments (scFvs). The optimization of scFv grafting resulted in Met-immunoliposomes with an average diameter of less than 200 nm, low polydispersity index (∼0.1), negative surface charge (<-10 mV), high Met drug loading (>150 mg g-1), and high affinity towards TROP 2 binding. Furthermore, Met-immunoliposomes were reproducible, and the scFv conjugation was stable in the presence of serum for five days. Their cellular uptake increased 4 folds in two-dimensional and 9 folds in three-dimensional TNBC models owing to the high affinity towards TROP 2 binding. Finally, it was observed that the therapeutic effect of Met in suppressing cancer cell growth and proliferation was superior when using anti-TROP 2 scFv-grafted Met-immunoliposomes, which completely stopped the spheroid growth and inhibited the expression of adenosine triphosphate. This study is one of the first reports to explore the combination of nanoparticle-based drug delivery systems to target the TROP 2 protein in TNBC, and to the best of our knowledge, this is the first report to specifically combine the use of scFvs with TROP 2 targeting to deliver therapeutics for TNBC treatment.
Collapse
Affiliation(s)
- Daiva Vozgirdaite
- UPR 4301 CBM, CNRS, NMNS Department, University of Tours, 37200 Tours, France.
| | | | | | - Emmanuel Douez
- UPR 4301 CBM, CNRS, NMNS Department, University of Tours, 37200 Tours, France.
- Pharmacy Department, University Hospital Center of Tours, 37200 Tours, France
| | - Louis Jolivet
- ISP UMR 1282, INRAE, BioMAP team, University of Tours, 37200 Tours, France
| | - Fanny Boursin
- ISP UMR 1282, INRAE, BioMAP team, University of Tours, 37200 Tours, France
| | - Igor Chourpa
- UPR 4301 CBM, CNRS, NMNS Department, University of Tours, 37200 Tours, France.
| | - Nicolas Aubrey
- ISP UMR 1282, INRAE, BioMAP team, University of Tours, 37200 Tours, France
| | - Katel Hervé-Aubert
- UPR 4301 CBM, CNRS, NMNS Department, University of Tours, 37200 Tours, France.
| |
Collapse
|
4
|
Ajay A, Wang H, Rezvani A, Savari O, Grubb BJ, McColl KS, Yoon S, Joseph PL, Kopp SR, Kresak AM, Peacock CD, Wildey GM, Lam M, Miyagi M, Kao HY, Dowlati A. Assessment of targets of antibody drug conjugates in SCLC. NPJ Precis Oncol 2025; 9:1. [PMID: 39748112 PMCID: PMC11696236 DOI: 10.1038/s41698-024-00784-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Antibody-drug conjugate (ADC) therapy has transformed treatment for several solid tumors, including small cell lung cancer (SCLC). However, significant challenges remain, including systemic toxicity, acquired resistance, and the lack of reliable biomarkers for patient selection. To enhance the effectiveness of ADC therapies in SCLC, we focused on target selection in this study by investigating the expression of ADC targets - SEZ6, DLL3, CD276, and TACSTD2 - in cell lines and patient samples. SEZ6 expression was significantly elevated in various SCLC transcriptional subtypes, particularly ASCL1, and exhibited gender-specific differences, being lower in women. DLL3 was primarily observed in the ASCL1 subtype, while CD276 showed high expression in non-neuroendocrine subtypes. TACSTD2 levels were generally low and attenuated in lymph nodes and brain metastases compared to primary tumors. Our findings underscore the importance of understanding target expression patterns to optimize ADC therapy and advance precision medicine in SCLC treatment.
Collapse
Affiliation(s)
- Abhishek Ajay
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Han Wang
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Ali Rezvani
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Omid Savari
- Department of Pathology, University Hospitals Cleveland Medical Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Brandon J Grubb
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Karen S McColl
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Suzy Yoon
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Peronne L Joseph
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Shelby R Kopp
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Adam M Kresak
- Department of Pathology, University Hospitals Cleveland Medical Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Craig D Peacock
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Gary M Wildey
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Minh Lam
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Masaru Miyagi
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Hung-Ying Kao
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Afshin Dowlati
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, and Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
5
|
Mihaylova R, Momekova D, Elincheva V, Momekov G. Immunoconjugates as an Efficient Platform for Drug Delivery: A Resurgence of Natural Products in Targeted Antitumor Therapy. Pharmaceuticals (Basel) 2024; 17:1701. [PMID: 39770542 PMCID: PMC11677665 DOI: 10.3390/ph17121701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The present review provides a detailed and comprehensive discussion on antibody-drug conjugates (ADCs) as an evolving new modality in the current therapeutic landscape of malignant diseases. The principle concepts of targeted delivery of highly toxic agents forsaken as stand-alone drugs are examined in detail, along with the biochemical and technological tools for their successful implementation. An extensive analysis of ADCs' major components is conducted in parallel with their function and impact on the stability, efficacy, safety, and resistance profiles of the immunoconjugates. The scope of the article covers the major classes of currently validated natural compounds used as payloads, with an emphasis on their structural and mechanistic features, natural origin, and distribution. Future perspectives in ADCs' design are thoroughly explored, addressing their inherent or emerging challenges and limitations. The survey also provides a comprehensive overview of the molecular rationale for active tumor targeting of ADC-based platforms, exploring the cellular biology and clinical relevance of validated tumor markers used as a "homing" mechanism in both hematological and solid tumor malignancies.
Collapse
Affiliation(s)
- Rositsa Mihaylova
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Denitsa Momekova
- Department “Pharmaceutical Technology and Biopharmaceutics”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria;
| | - Viktoria Elincheva
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Georgi Momekov
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| |
Collapse
|
6
|
Liu Y, Huang W, Saladin RJ, Hsu JC, Cai W, Kang L. Trop2-Targeted Molecular Imaging in Solid Tumors: Current Advances and Future Outlook. Mol Pharm 2024; 21:5909-5928. [PMID: 39537365 DOI: 10.1021/acs.molpharmaceut.4c00848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Trophoblast cell surface antigen 2 (Trop2), a transmembrane glycoprotein, plays a dual role in physiological and pathological processes. In healthy tissues, Trop2 facilitates development and orchestrates intracellular calcium signaling. However, its overexpression in numerous solid tumors shifts its function toward driving cell proliferation and metastasis, thus leading to a poor prognosis. The clinical relevance of Trop2 is underscored by its utility as both a biomarker for diagnostic imaging and a target for therapy. Notably, the U.S. Food and Drug Administration (FDA) has approved sacituzumab govitecan (SG), a novel Trop2-targeted agent, for treating triple-negative breast cancer (TNBC) and refractory urothelial cancer, highlighting the significance of Trop2 in clinical oncology. Molecular imaging, a powerful tool for visualizing and quantifying biological phenomena at the molecular and cellular levels, has emerged as a critical technique for studying Trop2. This approach encompasses various modalities, including optical imaging, positron emission tomography (PET), single photon emission computed tomography (SPECT), and targeted antibodies labeled with radioactive isotopes. Incorporating Trop2-targeted molecular imaging into clinical practice is vital for the early detection, prognostic assessment, and treatment planning of a broad spectrum of solid tumors. Our review captures the latest progress in Trop2-targeted molecular imaging, focusing on both diagnostic and therapeutic applications across diverse tumor types, including lung, breast, gastric, pancreatic, prostate, and cervical cancers, as well as salivary gland carcinomas. We critically evaluate the current state by examining the relevant applications, diagnostic accuracy, therapeutic efficacy, and inherent limitations. Finally, we analyze the challenges impeding widespread clinical application and offer insights into strategies for advancing the field, thereby guiding future research endeavors.
Collapse
Affiliation(s)
- Yongshun Liu
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Rachel J Saladin
- Departments of Radiology and Medical Physics, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
7
|
Astore S, Oneda E, Zaniboni A. A therapeutic algorithm guiding subsequent therapy selection after CDK4/6 inhibitors' failure: A review of current and investigational treatment for HR+/Her2- breast cancer. Crit Rev Oncol Hematol 2024; 204:104535. [PMID: 39433229 DOI: 10.1016/j.critrevonc.2024.104535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/23/2024] Open
Abstract
The first-line combination therapies utilizing cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) with endocrine therapy (ET) have significantly impacted the course of hormone receptor positive (HR+)/Human Epidermal Growth Factor Receptor 2 negative (HER2-) advanced breast cancer (ABC). However, resistance often emerges, leading to a molecularly different disease. Estrogen receptor one (ESR1) gene mutations, driving resistance to aromatase inhibitors (AIs), may guide the use of fulvestrant or emerging oral selective estrogen receptor degraders (SERDs) like elacestrant. The dynamic nature of ESR1 mutations suggests potential guidance for continuing CDK4/6i therapy beyond progression. Targeting mutations like breast cancer gene 1 and 2 (BRCA 1/2) with Poly (ADP-ribose) polymerase (PARP) inhibitors or the PI3K/AKT/mTOR pathway provides therapeutic options. The advent of antibody-drug conjugates (ADCs) like trastuzumab deruxtecan (T-DXd) and novel agents targeting Trophoblast cell surface antigen-2 (Trop-2) introduces further complexity, underscoring the need for early intervention targeting specific genomic alterations in metastatic BC.
Collapse
Affiliation(s)
- Serena Astore
- Medical Oncology Dept. Fondazione Poliambulanza, Brescia, Italy.
| | - Ester Oneda
- Medical Oncology Dept. Fondazione Poliambulanza, Brescia, Italy
| | | |
Collapse
|
8
|
Khoury K, Meisel JL, Yau C, Rugo HS, Nanda R, Davidian M, Tsiatis B, Chien AJ, Wallace AM, Arora M, Rozenblit M, Hershman DL, Zimmer A, Clark AS, Beckwith H, Elias AD, Stringer-Reasor E, Boughey JC, Nangia C, Vaklavas C, Omene C, Albain KS, Kalinsky KM, Isaacs C, Tseng J, Roussos Torres ET, Thomas B, Thomas A, Sanford A, Balassanian R, Ewing C, Yeung K, Sauder C, Sanft T, Pusztai L, Trivedi MS, Outhaythip A, Li W, Onishi N, Asare AL, Beineke P, Norwood P, Brown-Swigart L, Hirst GL, Matthews JB, Moore B, Fraser Symmans W, Price E, Beedle C, Perlmutter J, Pohlmann P, Shatsky RA, DeMichele A, Yee D, van 't Veer LJ, Hylton NM, Esserman LJ. Datopotamab-deruxtecan in early-stage breast cancer: the sequential multiple assignment randomized I-SPY2.2 phase 2 trial. Nat Med 2024; 30:3728-3736. [PMID: 39277671 DOI: 10.1038/s41591-024-03266-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/17/2024]
Abstract
Among the goals of patient-centric care are the advancement of effective personalized treatment, while minimizing toxicity. The phase 2 I-SPY2.2 trial uses a neoadjuvant sequential therapy approach in breast cancer to further these goals, testing promising new agents while optimizing individual outcomes. Here we tested datopotamab-deruxtecan (Dato-DXd) in the I-SPY2.2 trial for patients with high-risk stage 2/3 breast cancer. I-SPY2.2 uses a sequential multiple assignment randomization trial design that includes three sequential blocks of biologically targeted neoadjuvant treatment: the experimental agent(s) (block A), a taxane-based regimen tailored to the tumor subtype (block B) and doxorubicin-cyclophosphamide (block C). Patients are randomized into arms consisting of different investigational block A treatments. Algorithms based on magnetic resonance imaging and core biopsy guide treatment redirection after each block, including the option of early surgical resection in patients predicted to have a high likelihood of pathological complete response, the primary endpoint. There are two primary efficacy analyses: after block A and across all blocks for the six prespecified breast cancer subtypes (defined by clinical hormone receptor/human epidermal growth factor receptor 2 (HER2) status and/or the response-predictive subtypes). We report results of 103 patients treated with Dato-DXd. While Dato-DXd did not meet the prespecified threshold for success (graduation) after block A in any subtype, the treatment strategy across all blocks graduated in the hormone receptor-negative HER2-Immune-DNA repair deficiency- subtype with an estimated pathological complete response rate of 41%. No new toxicities were observed, with stomatitis and ocular events occurring at low grades. Dato-DXd was particularly active in the hormone receptor-negative/HER2-Immune-DNA repair deficiency- signature, warranting further investigation, and was safe in other subtypes in patients who followed the treatment strategy. ClinicalTrials.gov registration: NCT01042379 .
Collapse
Affiliation(s)
- Katia Khoury
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Christina Yau
- University of California San Francisco, San Francisco, CA, USA
| | - Hope S Rugo
- University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - A Jo Chien
- University of California San Francisco, San Francisco, CA, USA
| | | | - Mili Arora
- University of California Davis, Davis, CA, USA
| | | | | | | | - Amy S Clark
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | - Christos Vaklavas
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Coral Omene
- Cooperman Barnabas Medical Center, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Kathy S Albain
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL, USA
| | | | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | - Jennifer Tseng
- City of Hope Orange County Lennar Foundation Cancer Center, Orange County, CA, USA
| | | | | | | | | | | | - Cheryl Ewing
- University of California San Francisco, San Francisco, CA, USA
| | - Kay Yeung
- University of California San Diego, San Diego, CA, USA
| | | | | | | | | | | | - Wen Li
- University of California San Francisco, San Francisco, CA, USA
| | - Natsuko Onishi
- University of California San Francisco, San Francisco, CA, USA
| | - Adam L Asare
- University of California San Francisco, San Francisco, CA, USA
- Quantum Leap Healthcare Collaborative, San Francisco, CA, USA
| | - Philip Beineke
- Quantum Leap Healthcare Collaborative, San Francisco, CA, USA
| | - Peter Norwood
- Quantum Leap Healthcare Collaborative, San Francisco, CA, USA
| | | | - Gillian L Hirst
- University of California San Francisco, San Francisco, CA, USA
| | | | - Brian Moore
- Wake Forest University, Winston-Salem, NC, USA
| | | | - Elissa Price
- University of California San Francisco, San Francisco, CA, USA
| | - Carolyn Beedle
- University of California San Francisco, San Francisco, CA, USA
| | | | - Paula Pohlmann
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Douglas Yee
- University of Minnesota, Minneapolis, MN, USA
| | | | - Nola M Hylton
- University of California San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
9
|
Xi X, Wang Y, An G, Feng S, Zhu Q, Wu Z, Chen J, Zuo Z, Wang Q, Wang MW, Gu Y. A novel shark VNAR antibody-based immunotoxin targeting TROP-2 for cancer therapy. Acta Pharm Sin B 2024; 14:4806-4818. [PMID: 39664437 PMCID: PMC11628804 DOI: 10.1016/j.apsb.2024.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/11/2024] [Accepted: 07/15/2024] [Indexed: 12/13/2024] Open
Abstract
TROP-2, a tumor-associated antigen, has been implicated in the progression of various epithelial tumors. Due to its favorable expression profile, TROP-2 has emerged as a promising target for antibody-drug conjugates (ADCs) based anti-tumor therapies. Although ADCs have shown efficacy in cancer treatment, their application in solid tumors is hindered by their high molecular weight, poor tumor penetration, and release of cytotoxic molecules. Therefore, a recombinant immunotoxin was developed based on a shark-derived variable domain of immunoglobulin new antigen receptor (VNAR) antibody. VNARs are only one-tenth the size of IgG antibodies and possess remarkable tissue penetration capabilities and high stability. In this study, a shark VNAR phage display library was created, leading to the identification of shark VNAR-5G8 that targets TROP-2. VNAR-5G8 exhibited a high affinity and cellular internalization ability towards cells expressing high levels of TROP-2. Epitope analysis revealed that VNAR-5G8 recognizes a hidden epitope consisting of CRD and TY-1 on TROP-2. Subsequently, VNAR-5G8 was fused with a truncated form of Pseudomonas exotoxin (PE38) to create the recombinant immunotoxin (5G8-PE38), which exhibited significant anti-tumor activity in vitro and in vivo. Overall, this study highlights the promise of 5G8-PE38 as a valuable candidate for cancer therapy.
Collapse
Affiliation(s)
- Xiaozhi Xi
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao Marine Science and Technology Center, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Oncology Department, Shandong Second Provincial General Hospital, Jinan 250022, China
| | - Yanqing Wang
- Laboratory for Marine Drugs and Bioproducts of Qingdao Marine Science and Technology Center, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Guiqi An
- Laboratory for Marine Drugs and Bioproducts of Qingdao Marine Science and Technology Center, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Shitao Feng
- Laboratory for Marine Drugs and Bioproducts of Qingdao Marine Science and Technology Center, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Qiumei Zhu
- Laboratory for Marine Drugs and Bioproducts of Qingdao Marine Science and Technology Center, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhongqiu Wu
- Laboratory for Marine Drugs and Bioproducts of Qingdao Marine Science and Technology Center, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jin Chen
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Zhicheng Zuo
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Qiang Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan 250022, China
| | - Ming-Wei Wang
- Research Center for Deepsea Bioresources (Sanya), Hainan 572025, China
| | - Yuchao Gu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao Marine Science and Technology Center, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
10
|
Chen W, Zhang Y, Zhang L, Luo X, Yang X, Zhu Y, Wang G, Huang W, Zhang D, Zeng Y, Li R, Guo C, Wang J, Wu Z, Liu N, Zhang G. Intraoperative evaluation of tumor margins using a TROP2 near-infrared imaging probe to enable human breast-conserving surgery. Sci Transl Med 2024; 16:eado2461. [PMID: 39413161 DOI: 10.1126/scitranslmed.ado2461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Intraoperative surgical margin assessment remains a challenge during breast-conserving surgery. Here, we report a combined strategy of immuno-positron emission tomography (PET) for preoperative detection of breast cancer and guided assessment of margins in breast-conserving surgery through second near-infrared (NIR-II) fluorescence imaging of trophoblastic cell surface antigen 2 (TROP2). We demonstrated that the intensity of PET signals in the tumors was nearly five times higher than in normal breast tissue with a zirconium-89 tracer conjugated to sacituzumab govitecan (SG) in a mouse spontaneous breast cancer model, enabling the identification of tumors. We further generated a NIR-II probe of indocyanine green conjugated to SG (ICG-SG) and developed a rapid incubation imaging method for intraoperative margin assessment in a relevant time window for the operation workflow. The ICG-SG NIR-II fluorescence image guidance was first verified to remove tumors completely and accurately in mouse breast cancer models. Moreover, the rapid incubation imaging method was applied to distinguish benign and malignant breast lesions in samples from 26 patients with breast cancer. Therefore, we have developed both nuclide and optical probes targeting TROP2 for rapid and precise identification of tumor margins during breast-conserving surgery in humans.
Collapse
Affiliation(s)
- Weiling Chen
- Cancer Center and Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen 361101, China
| | - Yongqu Zhang
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Department of Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, China
| | - Lixin Zhang
- Cancer Center and Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen 361101, China
| | - Xiangjie Luo
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xia Yang
- Department of Nuclear Medicine, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Yuanyuan Zhu
- Cancer Center and Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen 361101, China
| | - Guimei Wang
- Department of Pathology, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Wenhe Huang
- Cancer Center and Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen 361101, China
| | - Deliang Zhang
- Department of Nuclear Medicine, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Yunzhu Zeng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou 515041, China
| | - Ronghui Li
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Department of Medical Oncology, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Cuiping Guo
- Department of Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jiazheng Wang
- Cancer Center and Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen 361101, China
| | - Zhao Wu
- Cancer Center and Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen 361101, China
| | - Na Liu
- Cancer Center and Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen 361101, China
| | - Guojun Zhang
- Cancer Center and Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen 361101, China
- Department of Breast Surgery, Yunnan Cancer Hospital, Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunan, Kunming 650118, China
- Cancer Research Center of Xiamen University, Xiamen 361101, China
| |
Collapse
|
11
|
Ralser DJ, Kiver V, Solomayer EF, Neeb C, Blohmer JU, Abramian AV, Maass N, Schütz F, Kolberg-Liedtke C, Müller C, Rambow AC. Impact of low HER2 expression on response to CDK4/6 inhibitor treatment in advanced HR + /HER2- breast cancer: a multicenter real-world data analysis. Arch Gynecol Obstet 2024:10.1007/s00404-024-07761-2. [PMID: 39373732 DOI: 10.1007/s00404-024-07761-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE CDK4/6 inhibitors (CDK4/6i) represent the first-line therapy approach of choice for patients with hormone receptor-positive, HER2-negative advanced breast cancer (HR + /HER-ABC). Approximately 50% of HR + /HER2-ABC displays low HER2 expression (HER2 low). Recent data emerging from the DESTINY-Breast04 trial demonstrated practice-changing efficacy of the antibody-drug conjugate trastuzumab deruxtecan (T-DXd) in patients with low HER2 expression. Here, we aimed to analyze the impact of low HER2 expression on CDK4/6i therapy response in a well-characterized multicenter HR + /HER-ABC cohort. METHODS Patients diagnosed with HR + /HER2-ABC who were treated with CDK4/6i in clinical routine between November 2016 and December 2020 at four certified German Breast Cancer Centers were retrospectively identified. The cohort was stratified according to graduation of positivity in HER2 immunohistochemistry (IHC; HER2 zero = IHC score 0 and HER2 low = IHC score 1 + , 2 + /fluorescence in situ hybridization negative). Subgroups were analyzed with regard to progression-free survival (PFS) following CDK4/6i initiation. FINDINGS The study cohort comprised n = 448 patients. For n = 311 patients, HER2 status from the metastatic site was available. n = 91 (29.3%) cases were HER2 zero and n = 220 cases (70.7%) were HER2 low. There was no significant difference in PFS between the two groups (PFS: 17 months versus 18 months, log-rank p = 0.42). Further, we examined the influence of HER2 expression changes between primary and metastatic tissue (n = 171; HER2 gain/HER2 loss/HER2 stable expression) on CDK4/6i treatment response. Again, there was no significant difference between these three groups, respectively (PFS: 16 months versus 13 months versus 17 months, log-rank p = 0.86). CONCLUSIONS In our analysis, HER2 status did not have a significant impact on treatment response to CDK4/6i.
Collapse
Affiliation(s)
- Damian J Ralser
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Verena Kiver
- Department of Gynecology With Breast Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Erich-Franz Solomayer
- Department of Gynecology, Obstetrics & Reproductive Medicine, Saarland University Medical Center, Homburg, Saar, Germany
| | - Caroline Neeb
- Department of Gynecology With Breast Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jens-Uwe Blohmer
- Department of Gynecology With Breast Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Alina V Abramian
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, University Medical Center Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Nicolai Maass
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | - Florian Schütz
- Department of Gynecology and Obstetrics, Diakonissen-Stiftungs-Krankenhaus Speyer, Speyer, Germany
| | | | - Carolin Müller
- Department of Gynecology, Obstetrics & Reproductive Medicine, Saarland University Medical Center, Homburg, Saar, Germany
- Department of Anesthesiology, Outcomes Research Consortium, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Anna-Christina Rambow
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| |
Collapse
|
12
|
Gao X, Bu T, Wang W, Xu Y. Comparative Analysis and Future Prospects of Human Epidermal Growth Factor Receptor 2 (HER2) and Trophoblast Cell-Surface Antigen 2 (Trop-2) Targeted Antibody-Drug Conjugates in Breast Cancer Treatment. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:621-630. [PMID: 39310781 PMCID: PMC11416103 DOI: 10.2147/bctt.s480796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024]
Abstract
Breast cancer remains the most prevalent malignancy among women globally, presenting significant challenges in therapeutic strategies due to tumor heterogeneity, drug resistance, and adverse side effects. Recent advances in targeted therapies, particularly antibody-drug conjugates (ADCs), have shown promise in addressing these challenges by selectively targeting tumor cells while sparing normal tissues. This study provides a comprehensive analysis of two innovative ADCs targeting HER2 and Trop-2, which are critical markers in various breast cancer subtypes. These conjugates combine potent cytotoxic drugs with specific antibodies, leveraging the antigens' differential expression to enhance therapeutic efficacy and reduce systemic toxicity. Our comparative analysis highlights the clinical applications, efficacy, and safety profiles of these ADCs, drawing on data from recent clinical trials. In addition, the paper discusses the potential of these ADCs in treating other types of cancers where HER2 and Trop-2 are expressed, as well as the toxicity risks associated with targeting these antigens in normal cells. Additionally, the paper discusses novel synthetic drugs that show potential in preclinical models, focusing on their mechanisms of action and therapeutic advantages over traditional chemotherapy. The findings underscore the transformative impact of targeted ADCs in breast cancer treatment, noting significant advancements in patient outcomes and management of side effects. However, ongoing issues such as resistance mechanisms and long-term safety remain challenges. The conclusion offers a forward-looking perspective on potential improvements and the future trajectory of ADC research. This study not only elucidates the current landscape of ADCs in breast cancer but also sets the stage for the next generation of oncological therapeutics. This study not only elucidates the current landscape of ADCs in breast cancer but also sets the stage for the next generation of oncological therapeutics, with particular attention to their broader applications and associated risks.
Collapse
Affiliation(s)
- Xiaojuan Gao
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, Gansu Province, People’s Republic of China
| | - Tiansheng Bu
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, Gansu Province, People’s Republic of China
| | - Wenying Wang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, Gansu Province, People’s Republic of China
| | - Ying Xu
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, Gansu Province, People’s Republic of China
| |
Collapse
|
13
|
Wang J, Tong Z, Tan Y, Shi Y, Wu Y, Zhou Q, Xing X, Chen X, Qiu F, Ma F. Phase 1a study of ESG401, a Trop2 antibody-drug conjugate, in patients with locally advanced/metastatic solid tumors. Cell Rep Med 2024; 5:101707. [PMID: 39216478 PMCID: PMC11524954 DOI: 10.1016/j.xcrm.2024.101707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/14/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
This phase 1a study assesses ESG401 in patients with heavily pretreated locally advanced or metastatic solid tumors, focusing on metastatic breast cancer. Forty patients are enrolled: three experience dose-limiting toxicities, establishing the maximum tolerated dose at 16 mg/kg on days 1, 8, and 15 of a 28-day cycle. The most common grade ≥3 treatment-related adverse events are neutropenia and leukopenia. Among 38 efficacy-evaluable patients, the objective response rate (ORR) is 34.2%, the disease control rate (DCR) is 65.8%, and the clinical benefit rate (CBR) is 50.0% (including stable disease for at least 6 months). The median progression-free survival is 5.1 months, and the median duration of response is 6.3 months. In patients receiving therapeutically relevant doses, the ORR, DCR, and CBR are 40.6%, 75.0%, and 56.3%, respectively. ESG401 demonstrates a favorable safety profile and promising antitumor activity in this heavily treated population. The trial is registered at ClinicalTrials.gov (NCT04892342).
Collapse
Affiliation(s)
- Jiani Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongsheng Tong
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yinuo Tan
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yehui Shi
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yun Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Zhou
- Shanghai Escugen Biotechnology Co., Ltd., Shanghai, China
| | - Xiaoyan Xing
- Shanghai Escugen Biotechnology Co., Ltd., Shanghai, China
| | - Xiaomei Chen
- Shanghai Escugen Biotechnology Co., Ltd., Shanghai, China
| | - Fuming Qiu
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Mueller C, Davis JB, Espina V. Protein biomarkers for subtyping breast cancer and implications for future research: a 2024 update. Expert Rev Proteomics 2024; 21:401-416. [PMID: 39474929 DOI: 10.1080/14789450.2024.2423625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Breast cancer subtyping is used clinically for diagnosis, prognosis, and treatment decisions. Subtypes are categorized by cell of origin, histomorphology, gene expression signatures, hormone receptor status, and/or protein levels. Categorizing breast cancer based on gene expression signatures aids in assessing a patient's recurrence risk. Protein biomarkers, on the other hand, provide functional data for selecting therapies for primary and recurrent tumors. We provide an update on protein biomarkers in breast cancer subtypes and their application in prognosis and therapy selection. AREAS COVERED Protein pathways in breast cancer subtypes are reviewed in the context of current protein-targeted treatment options. PubMed, Science Direct, Scopus, and Cochrane Library were searched for relevant studies between 2017 and 17 August 2024. EXPERT OPINION Post-translationally modified proteins and their unmodified counterparts have become clinically useful biomarkers for defining breast cancer subtypes from a therapy perspective. Tissue heterogeneity influences treatment outcomes and disease recurrence. Spatial profiling has revealed complex cellular subpopulations within the breast tumor microenvironment. Deciphering the functional relationships between and within tumor clonal cell populations will further aid in defining breast cancer subtypes and create new treatment paradigms for recurrent, drug resistant, and metastatic disease.
Collapse
Affiliation(s)
- Claudius Mueller
- Laboratory and Bioinformatics Department, Ignite Proteomics, Golden, CO, USA
| | - Justin B Davis
- Laboratory and Bioinformatics Department, Ignite Proteomics, Golden, CO, USA
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| |
Collapse
|
15
|
Jaime-Casas S, Barragan-Carrillo R, Tripathi A. Antibody-drug conjugates in solid tumors: a new frontier. Curr Opin Oncol 2024; 36:421-429. [PMID: 39007183 DOI: 10.1097/cco.0000000000001064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) are designed to carry cytotoxic payloads and deliver them to specific molecular targets within tumor cells. Several ADCs are already approved with many more in development across several disease types. In this review, we will provide an overview of the ADCs currently approved and those under investigation in solid tumors. RECENT FINDINGS Currently there are dozens of ADCs under clinical study evaluation of a variety of solid tumors, and preliminary results are promising. Multiple ADCs have received regulatory approval in disease such as breast cancer, non-small cell lung cancer, and bladder cancer. While some are approved in biomarker selected settings with disease specific indication (e.g. breast cancer), others have been approved irrespective of biomarker expression (urothelial carcinoma) and pan-cancer indications in biomarker selected patients (HER2 3+ expression). SUMMARY Cytotoxic chemotherapy has been the mainstay of systemic treatment for patients with various solid tumors. ADCs offer the advantage of carrying the cytotoxic payload onto a specific molecular receptor, thereby inducing a more selective response. Optimizing selection of target antigen, payload delivery and investigating biomarkers of response will be crucial for further expanding the therapeutic benefit of ADCs across solid tumors.
Collapse
Affiliation(s)
- Salvador Jaime-Casas
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | | | | |
Collapse
|
16
|
Pinto A, Guarini C, Giampaglia M, Sanna V, Melaccio A, Lanotte L, Santoro AN, Pini F, Cusmai A, Giuliani F, Gadaleta-Caldarola G, Fedele P. Synergizing Immunotherapy and Antibody-Drug Conjugates: New Horizons in Breast Cancer Therapy. Pharmaceutics 2024; 16:1146. [PMID: 39339183 PMCID: PMC11435286 DOI: 10.3390/pharmaceutics16091146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
The advent of immunotherapy and antibody-drug conjugates (ADCs) have revolutionized breast cancer treatment, offering new hope to patients. However, challenges, such as resistance and limited efficacy in certain cases, remain. Recently, the combination of these therapies has emerged as a promising approach to address these challenges. ADCs play a crucial role by delivering cytotoxic agents directly to breast cancer cells, minimizing damage to healthy tissue and enhancing the tumor-killing effect. Concurrently, immunotherapies harness the body's immune system to recognize and eliminate cancer cells. This integration offers potential to overcome resistance mechanisms and significantly improve therapeutic outcomes. This review explores the rationale behind combining immunotherapies with ADCs, recent advances in this field, and the potential implications for breast cancer treatment.
Collapse
Affiliation(s)
- Antonello Pinto
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| | - Chiara Guarini
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| | | | - Valeria Sanna
- Oncology Unit, "Ospedale Civile Santissima Annunziata" Hospital, 07100 Sassari, Italy
| | | | - Laura Lanotte
- Oncology Unit, "Mons. Dimiccoli" Hospital, 70051 Barletta, Italy
| | | | - Francesca Pini
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| | - Antonio Cusmai
- "Don Tonino Bello", I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy
| | | | | | - Palma Fedele
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| |
Collapse
|
17
|
Rossi V, Turati A, Rosato A, Carpanese D. Sacituzumab govitecan in triple-negative breast cancer: from bench to bedside, and back. Front Immunol 2024; 15:1447280. [PMID: 39211043 PMCID: PMC11357913 DOI: 10.3389/fimmu.2024.1447280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Triple-negative breast cancer (TNBC) represents a major therapeutic challenge due to its heterogeneous and aggressive phenotype, and limited target-specific treatment options. The trophoblast cell surface antigen (Trop-2), a transmembrane glycoprotein overexpressed in various cancers, has emerged as a promising target for TNBC. Sacituzumab govitecan (SG), an antibody-drug conjugate (ADC) that targets Trop-2, has recently entered treatment algorithms for advanced and metastatic TNBC, independently from Trop-2 expression status, with manageable toxicity. Despite the impressive results, questions remain unsolved regarding its efficacy, safety profile, and Trop-2 biological role in cancer. Currently, Trop-2 cannot be designated as a predictive biomarker in SG treatment, albeit its expression correlates with disease outcome, yet its levels are not uniform across all TNBCs. Additionally, data regarding Trop-2 expression variations in primary and metastatic sites, and its interplay with other biomarkers are still ambiguous but mandatory in light of future applications of SG in other indications and settings. This poses the questions of a careful evaluation of the efficacy and toxicity profile of SG in such early stages of disease, and in personalized and combinatorial strategies. Research and clinical data are mandatory to address SG drawbacks and minimize its benefits, to realize its full potential as therapeutic agent in different epithelial tumors.
Collapse
Affiliation(s)
- Valentina Rossi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| | - Alessandra Turati
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Debora Carpanese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| |
Collapse
|
18
|
Udvorková N, Fekiačová A, Majtánová K, Mego M, Kučerová L. Antibody-drug conjugates as a novel therapeutic modality to treat recurrent refractory germ cell tumors. Am J Physiol Cell Physiol 2024; 327:C362-C371. [PMID: 38912730 DOI: 10.1152/ajpcell.00200.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
This review provides a rationale for using the Food and Drug Administration (FDA)-approved antibody-drug conjugates (ADCs) for implementing as therapy in recurrent refractory germ cell tumors similar to their position in the treatment of other types of chemoresistant solid tumors. Germ cell tumors (GCTs) originate from germ cells; they most frequently develop in ovaries or in the testes, while being the most common type of malignancy in young men. GCTs are very sensitive to cisplatin-based chemotherapy, but therapeutic resistance occurs in a considerable number of cases, which is associated with disease recurrence and poor patient prognosis. ADCs are a novel type of targeted antitumor agents that combine tumor antigen-specific monoclonal antibodies with chemically linked chemotherapeutic drugs (payload) exerting a cytotoxic effect. Several FDA-approved ADCs use as targeting moieties the antigens that are also detected in the GCTs, offering a benefit of this type of targeted therapy even for patients with relapsed/refractory testicular GCTs (rrTGCT) unresponsive to standard chemotherapy.
Collapse
Affiliation(s)
- Natália Udvorková
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Cancer Research Institute, Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Adriana Fekiačová
- Cancer Research Institute, Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Genetics, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Kristína Majtánová
- Cancer Research Institute, Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
- Translational Research Unit, National Cancer Institute and the 2nd Oncology Clinic of Medical Faculty, Comenius University, Bratislava, Slovakia
| | - Michal Mego
- Translational Research Unit, National Cancer Institute and the 2nd Oncology Clinic of Medical Faculty, Comenius University, Bratislava, Slovakia
| | - Lucia Kučerová
- Cancer Research Institute, Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
- Translational Research Unit, National Cancer Institute and the 2nd Oncology Clinic of Medical Faculty, Comenius University, Bratislava, Slovakia
| |
Collapse
|
19
|
Bardia A, Krop IE, Kogawa T, Juric D, Tolcher AW, Hamilton EP, Mukohara T, Lisberg A, Shimizu T, Spira AI, Tsurutani J, Damodaran S, Papadopoulos KP, Greenberg J, Kobayashi F, Zebger-Gong H, Wong R, Kawasaki Y, Nakamura T, Meric-Bernstam F. Datopotamab Deruxtecan in Advanced or Metastatic HR+/HER2- and Triple-Negative Breast Cancer: Results From the Phase I TROPION-PanTumor01 Study. J Clin Oncol 2024; 42:2281-2294. [PMID: 38652877 PMCID: PMC11210948 DOI: 10.1200/jco.23.01909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/09/2024] [Accepted: 02/08/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Datopotamab deruxtecan (Dato-DXd) is an antibody-drug conjugate consisting of a humanized antitrophoblast cell-surface antigen 2 (TROP2) monoclonal antibody linked to a potent, exatecan-derived topoisomerase I inhibitor payload via a plasma-stable, selectively cleavable linker. PATIENTS AND METHODS TROPION-PanTumor01 (ClinicalTrials.gov identifier: NCT03401385) is a phase I, dose-escalation, and dose-expansion study evaluating Dato-DXd in patients with previously treated solid tumors. The primary study objective was to assess the safety and tolerability of Dato-DXd. Secondary objectives included evaluation of antitumor activity and pharmacokinetics. Results from patients with advanced/metastatic hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) breast cancer (BC) or triple-negative BC (TNBC) are reported. RESULTS At data cutoff (July 22, 2022), 85 patients (HR+/HER2- BC = 41, and TNBC = 44) had received Dato-DXd. The objective response rate by blinded independent central review was 26.8% (95% CI, 14.2 to 42.9) and 31.8% (95% CI, 18.6 to 47.6) for patients with HR+/HER2- BC and TNBC, respectively. The median duration of response was not evaluable in the HR+/HER2- BC cohort and 16.8 months in the TNBC cohort. The median progression-free survival in patients with HR+/HER2- BC and TNBC was 8.3 and 4.4 months, respectively. All-cause treatment-emergent adverse events (TEAEs; any grade, grade ≥3) were observed in 100% and 41.5% of patients with HR+/HER2- BC and 100% and 52.3% of patients with TNBC. Stomatitis was the most common TEAE (any grade, grade ≥3) in both HR+/HER2- BC (82.9%, 9.8%) and TNBC (72.7%, 11.4%) cohorts. CONCLUSION In patients with heavily pretreated advanced HR+/HER2- BC and TNBC, Dato-DXd demonstrated promising clinical activity and a manageable safety profile. Dato-DXd is currently being evaluated in phase III studies.
Collapse
MESH Headings
- Humans
- Female
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/pathology
- Middle Aged
- Aged
- Immunoconjugates/therapeutic use
- Immunoconjugates/adverse effects
- Immunoconjugates/pharmacokinetics
- Adult
- Receptor, ErbB-2/metabolism
- Camptothecin/analogs & derivatives
- Camptothecin/therapeutic use
- Receptors, Estrogen/metabolism
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Receptors, Progesterone/metabolism
- Antigens, Neoplasm
- Cell Adhesion Molecules/metabolism
- Trastuzumab
Collapse
Affiliation(s)
- Aditya Bardia
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Ian E. Krop
- Yale Cancer Center, New Haven, CT
- Dana-Farber Cancer Institute, Boston, MA
| | - Takahiro Kogawa
- Department of Advanced Medical Development, Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Dejan Juric
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Anthony W. Tolcher
- South Texas Accelerated Research Therapeutics, San Antonio, TX
- NEXT Oncology, San Antonio, TX
- Texas Oncology, San Antonio, TX
| | - Erika P. Hamilton
- Sarah Cannon Research Institute, Nashville, TN
- Tennessee Oncology, PLLC, Nashville, TN
| | - Toru Mukohara
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Aaron Lisberg
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Toshio Shimizu
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Pulmonary Medicine and Medical Oncology, Wakayama Medical University Hospital, Wakayama, Japan
| | | | - Junji Tsurutani
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Senthil Damodaran
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Jonathan Greenberg
- Global Oncology Clinical Development, Daiichi Sankyo, Inc, Basking Ridge, NJ
- Global Oncology Clinical Development, Daiichi Sankyo Europe GmbH, Munich, Germany
| | | | - Hong Zebger-Gong
- Global Oncology Clinical Development, Daiichi Sankyo Europe GmbH, Munich, Germany
| | - Rie Wong
- Global Oncology Clinical Development, Daiichi Sankyo, Co, Ltd, Tokyo, Japan
| | - Yui Kawasaki
- Global Oncology Clinical Development, Daiichi Sankyo, Inc, Basking Ridge, NJ
| | | | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
20
|
Parit S, Manchare A, Gholap AD, Mundhe P, Hatvate N, Rojekar S, Patravale V. Antibody-Drug Conjugates: A promising breakthrough in cancer therapy. Int J Pharm 2024; 659:124211. [PMID: 38750981 DOI: 10.1016/j.ijpharm.2024.124211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
Antibody-drug conjugates (ADCs) provide effective cancer treatment through the selective delivery of cytotoxic payloads to the cancer cells. They offer unparalleled precision and specificity in directing drugs to cancer cells while minimizing off-target effects. Despite several advantages, there is a requirement for innovations in the molecular design of ADC owing to drug resistance, cancer heterogeneity along the adverse effects of treatment. The review critically analyses ADC function mechanisms, unraveling the intricate interplay between antibodies, linkers, and payloads in facilitating targeted drug delivery to cancer cells. The article also highlights notable advancements in antibody engineering, which aid in creating highly selective and potent ADCs. Additionally, the review details significant progress in clinical ADC development with an in-depth examination of pivotal trials and approved formulations. Antibody Drug Conjugates (ADCs) are a ground-breaking approach to targeted drug delivery, especially in cancer treatment. They offer unparalleled precision and specificity in directing drugs to cancer cells while minimizing off-target effects. This review provides a comprehensive examination of the current state of ADC development, covering their design, mechanisms of action, and clinical applications. The article emphasizes the need for greater precision in drug delivery and explains why ADCs are necessary.
Collapse
Affiliation(s)
- Swapnali Parit
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Ajit Manchare
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Prashant Mundhe
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Navnath Hatvate
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Satish Rojekar
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
21
|
Rajadurai P, Ravindran S, Lee BR, Md Pauzi SH, Chiew SF, Teoh KH, S Raja Gopal N, Md Yusof M, Yip CH. Consensus Guidelines on Human Epidermal Growth Factor Receptor 2 (HER2)-Low Testing in Breast Cancer in Malaysia. Cancers (Basel) 2024; 16:2325. [PMID: 39001387 PMCID: PMC11240573 DOI: 10.3390/cancers16132325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/18/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Breast cancer is one of the most common cancers in Malaysia. Recently, a new nomenclature was introduced for breast cancers with human epidermal growth factor receptor 2 (HER2) immunohistochemistry (IHC) 1+, or 2+ with negative in situ hybridization (ISH), i.e., HER2-low breast cancer. In current clinical practice, these breast cancers are reported as HER2-negative. Clinical trials have shown that HER2-low breast cancer benefits from targeted therapy with anti-HER2 antibody-drug conjugates. Unfortunately, various challenges and obstacles are faced by local pathologists in HER2 testing, which may jeopardize the standard of care for patients with HER2-low breast cancer. This consensus guideline aims to elucidate standard practices pertaining to HER2 testing and HER2-low interpretation in Malaysia. Topics discussed among a panel of local experts include tissue sampling and handling, assay and antibody selection, result interpretation and reporting, and quality assurance. Practice recommendations made in this consensus guideline reflect current international guidelines and, where appropriate, adapted to the Malaysian landscape.
Collapse
Affiliation(s)
- Pathmanathan Rajadurai
- Subang Jaya Medical Centre, Subang Jaya 47500, Malaysia
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya 47500, Malaysia
- Department of Pathology, University of Malaya Medical Centre, Lembah Pantai, Kuala Lumpur 59100, Malaysia
| | - Sarala Ravindran
- Premier Integrated Labs, Pantai Hospital Kuala Lumpur, Kuala Lumpur 59100, Malaysia
| | | | - Suria Hayati Md Pauzi
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur 56000, Malaysia
| | - Seow Fan Chiew
- Department of Pathology, University of Malaya Medical Centre, Lembah Pantai, Kuala Lumpur 59100, Malaysia
| | - Kean Hooi Teoh
- Sunway Medical Centre, Bandar Sunway, Subang Jaya 47500, Malaysia
| | | | | | - Cheng Har Yip
- Subang Jaya Medical Centre, Subang Jaya 47500, Malaysia
| |
Collapse
|
22
|
Iyer K, Ivanov J, Tenchov R, Ralhan K, Rodriguez Y, Sasso JM, Scott S, Zhou QA. Emerging Targets and Therapeutics in Immuno-Oncology: Insights from Landscape Analysis. J Med Chem 2024; 67:8519-8544. [PMID: 38787632 PMCID: PMC11181335 DOI: 10.1021/acs.jmedchem.4c00568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
In the ever-evolving landscape of cancer research, immuno-oncology stands as a beacon of hope, offering novel avenues for treatment. This study capitalizes on the vast repository of immuno-oncology-related scientific documents within the CAS Content Collection, totaling over 350,000, encompassing journals and patents. Through a pioneering approach melding natural language processing with the CAS indexing system, we unveil over 300 emerging concepts, depicted in a comprehensive "Trend Landscape Map". These concepts, spanning therapeutic targets, biomarkers, and types of cancers among others, are hierarchically organized into eight major categories. Delving deeper, our analysis furnishes detailed quantitative metrics showcasing growth trends over the past three years. Our findings not only provide valuable insights for guiding future research endeavors but also underscore the merit of tapping the vast and unparalleled breadth of existing scientific information to derive profound insights.
Collapse
Affiliation(s)
| | - Julian Ivanov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Janet M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Sabina Scott
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
23
|
Gion M, García-Mosquera JJ, Pérez-García JM, Peg V, Ruiz-Borrego M, Stradella A, Bermejo B, Guerrero JA, López-Montero L, Mancino M, Rodríguez-Morató J, Antonarelli G, Sampayo-Cordero M, Llombart-Cussac A, Cortés J. Correlation between trophoblast cell-surface antigen-2 (Trop-2) expression and pathological complete response in patients with HER2-positive early breast cancer treated with neoadjuvant docetaxel, carboplatin, trastuzumab, and pertuzumab. Breast Cancer Res Treat 2024; 205:589-598. [PMID: 38456970 DOI: 10.1007/s10549-024-07292-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/08/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE The prognostic and predictive role of trophoblast cell-surface antigen-2 (Trop-2) overexpression in human epidermal growth factor receptor 2-positive (HER2-positive) breast cancer is currently unknown. We retrospectively analyzed Trop-2 expression and its correlation with clinicopathologic features and pathological complete response (pCR) in HER2-positive early breast cancer (EBC) patients treated with neoadjuvant docetaxel, carboplatin, trastuzumab, and pertuzumab in the PHERGain study. METHODS Trop-2 expression at baseline was determined in formalin-fixed, paraffin-embedded primary tumor biopsies by immunohistochemistry and was first classified into expressing (Trop-2-positive) or not-expressing (Trop-2-negative) tumors. Then, it was classified by histochemical score (H-score) according to its intensity into low (0-9), intermediate (10-49), and high (≥ 50). The association between clinicopathologic features, pCR, and Trop-2 expression was performed with Fisher's exact test. RESULTS Forty-one patients with tissue evaluable for Trop-2 expression were included, with 28 (68.3%) Trop-2-positive tumors. Overall, 17 (41.46%), 14 (34.15%), and 10 (24.40%) tumors were classified as low, intermediate, and high, respectively. Trop-2 expression was significantly associated with decreased pCR rates (50.0% vs. 92.3%; odds ratio [OR] 0.05; 95% CI, 0.002-0.360]; p adjusted = 0.01) but was not correlated with any clinicopathologic features (p ≥ 0.05). Tumors with the highest Trop-2 H-score were less likely to obtain a pCR (OR 0.03; 95% CI, 0.001-0.290, p adjusted < 0.01). This association was confirmed in univariate and multivariate regression analyses. CONCLUSION These findings suggest a potential role of Trop-2 expression as a biomarker of resistance to neoadjuvant chemotherapy plus dual HER2 blockade and may become a strategic target for future combinations in HER2-positive EBC patients.
Collapse
Affiliation(s)
- María Gion
- Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Juan José García-Mosquera
- Dr. Rosell Oncology Institute (IOR), Dexeus University Hospital, Quironsalud Group, Barcelona, Spain
| | - José Manuel Pérez-García
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Vicente Peg
- Department of Pathology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Agostina Stradella
- Institut Català d'Oncologia L'Hospitalet, Hospitalet de Llobregat, Barcelona, Spain
| | - Begoña Bermejo
- Hospital Clínico Universitario de Valencia, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - José Antonio Guerrero
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Laura López-Montero
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Mario Mancino
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - José Rodríguez-Morató
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Gabriele Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Miguel Sampayo-Cordero
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Antonio Llombart-Cussac
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain.
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA.
- Hospital Arnau de Vilanova, Valencia, Spain.
- Universidad Católica de Valencia, Valencia, Spain.
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain.
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain.
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA.
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain.
| |
Collapse
|
24
|
Schipilliti FM, Drittone D, Mazzuca F, La Forgia D, Guven DC, Rizzo A. Datopotamab deruxtecan: A novel antibody drug conjugate for triple-negative breast cancer. Heliyon 2024; 10:e28385. [PMID: 38560142 PMCID: PMC10981107 DOI: 10.1016/j.heliyon.2024.e28385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Triple negative breast cancer (TNBC) represents the breast cancer subtype with least favorable outcome because of the lack of effective treatment options and its molecular features. Recently, ADCs have dramatically changed the breast cancer treatment landscape; the anti-TROP2 ADC Sacituzumab Govitecan has been approved for treatment of previously treated, metastatic TNBC patients. The novel ADC Datopotecan-deruxtecan (Dato-DXd) has recently shown encouraging results for TNBC. In the current paper, we summarize and discuss available data regarding this TROP-2 directed agent mechanism of action and pharmacologic activity, we describe first results on efficacy and safety of the drug and report characteristics, inclusion criteria and endpoints of the main ongoing clinical trials.
Collapse
Affiliation(s)
| | - Denise Drittone
- Oncological Department, Sant'Andrea Hospital, University Sapienza in Rome, Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, Sapienza University, Oncology Unit, Azienda Ospedialiera Universitaria Sant'Andrea, Rome, Italy
| | | | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100, Sihhiye, Ankara, Turkey
| | | |
Collapse
|
25
|
Heist RS, Sands J, Bardia A, Shimizu T, Lisberg A, Krop I, Yamamoto N, Kogawa T, Al-Hashimi S, Fung SSM, Galor A, Pisetzky F, Basak P, Lau C, Meric-Bernstam F. Clinical management, monitoring, and prophylaxis of adverse events of special interest associated with datopotamab deruxtecan. Cancer Treat Rev 2024; 125:102720. [PMID: 38502995 DOI: 10.1016/j.ctrv.2024.102720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/07/2024] [Accepted: 03/10/2024] [Indexed: 03/21/2024]
Abstract
Antibody drug conjugates (ADCs) are an emerging class of treatments designed to improve efficacy and decrease toxicity compared with other systemic therapies through the selective delivery of cytotoxic agents to tumor cells. Datopotamab deruxtecan (Dato-DXd) is a novel ADC comprising a topoisomerase I inhibitor payload and a monoclonal antibody directed to trophoblast cell-surface antigen 2 (TROP2), a protein that is broadly expressed in several types of solid tumors. Dato-DXd is being investigated across multiple solid tumor indications. In the ongoing, first-in-human TROPION-PanTumor01 phase I study (ClinicalTrials.gov: NCT03401385), encouraging and durable antitumor activity and a manageable safety profile was demonstrated in patients with advanced/metastatic hormone receptor-positive/human epidermal growth factor receptor2-negative breast cancer (HR+/HER2- BC), triple-negative breast cancer (TNBC), and non-small cell lung cancer (NSCLC). Improved understanding of the adverse events (AEs) that are associated with Dato-DXd and their optimal management is essential to ensure safe and successful administration. Interstitial lung disease/pneumonitis, infusion-related reactions, oral mucositis/stomatitis, and ocular surface events have been identified as AEs of special interest (AESIs) for which appropriate prevention, monitoring, and management is essential. This article summarizes the incidence of AESIs among patients with HR+/HER2- BC, TNBC, and NSCLC reported in TROPION-PanTumor01. We report our recommendations for AESI prophylaxis, early detection, and management, using experience gained from treating AESIs that occur with Dato-DXd in clinical trials.
Collapse
Affiliation(s)
- Rebecca S Heist
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Harvard University, Boston, MA, USA.
| | - Jacob Sands
- Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Aditya Bardia
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Toshio Shimizu
- Department of Pulmonary Medicine and Medical Oncology, Wakayama Medical University Hospital, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Aaron Lisberg
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ian Krop
- Yale Cancer Center, New Haven, CT, USA
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Kogawa
- Department of Advanced Medical Development, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Saba Al-Hashimi
- Department of Ophthalmology, UCLA Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Simon S M Fung
- Department of Ophthalmology, UCLA Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Anat Galor
- Bascom Palmer Eye Institute, Miller School of Medicine, University of Miami, FL, USA; Research Services, Miami Veterans Affairs Medical Center, Miami, FL, USA
| | - Francesca Pisetzky
- Clinical Safety and Pharmacovigilence, Daiichi Sankyo, Inc., Schiphol-Rijk, The Netherlands
| | - Priyanka Basak
- Clinical Safety and Pharmacovigilance, Daiichi Sankyo, Inc., Basking Ridge, NJ, USA
| | - Cindy Lau
- Clinical Safety and Pharmacovigilance, Daiichi Sankyo, Inc., Basking Ridge, NJ, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| |
Collapse
|
26
|
Mustafa M, Abbas K, Alam M, Ahmad W, Moinuddin, Usmani N, Siddiqui SA, Habib S. Molecular pathways and therapeutic targets linked to triple-negative breast cancer (TNBC). Mol Cell Biochem 2024; 479:895-913. [PMID: 37247161 DOI: 10.1007/s11010-023-04772-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/18/2023] [Indexed: 05/30/2023]
Abstract
Cancer is a group of diseases characterized by uncontrolled cellular growth, abnormal morphology, and altered proliferation. Cancerous cells lose their ability to act as anchors, allowing them to spread throughout the body and infiltrate nearby cells, tissues, and organs. If these cells are not identified and treated promptly, they will likely spread. Around 70% of female breast cancers are caused by a mutation in the BRCA gene, specifically BRCA1. The absence of progesterone, oestrogen and HER2 receptors (human epidermal growth factor) distinguishes the TNBC subtype of breast cancer. There were approximately 6,85,000 deaths worldwide and 2.3 million new breast cancer cases in women in 2020. Breast cancer is the most common cancer globally, affecting 7.8 million people at the end of 2020. Compared to other cancer types, breast cancer causes more women to lose disability-adjusted life years (DALYs). Worldwide, women can develop breast cancer at any age after puberty, but rates increase with age. The maintenance of mammary stem cell stemness is disrupted in TNBC, governed by signalling cascades controlling healthy mammary gland growth and development. Interpreting these essential cascades may facilitate an in-depth understanding of TNBC cancer and the search for an appropriate therapeutic target. Its treatment remains challenging because it lacks specific receptors, which renders hormone therapy and medications ineffective. In addition to radiotherapy, numerous recognized chemotherapeutic medicines are available as inhibitors of signalling pathways, while others are currently undergoing clinical trials. This article summarizes the vital druggable targets, therapeutic approaches, and strategies associated with TNBC.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Kashif Abbas
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | - Moinuddin
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Nazura Usmani
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Shahid Ali Siddiqui
- Department of Radiotherapy, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India.
| |
Collapse
|
27
|
Mertens RB, Makhoul EP, Li X, Dadmanesh F. Comparative expression of trophoblast cell-surface antigen 2 (TROP2) in the different molecular subtypes of invasive breast carcinoma: An immunohistochemical study of 94 therapy-naive primary breast tumors. Ann Diagn Pathol 2024; 68:152226. [PMID: 37995412 DOI: 10.1016/j.anndiagpath.2023.152226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Sacituzumab govitecan, targeting trophoblast cell-surface antigen 2 (TROP2), is approved for the treatment of triple-negative and hormone receptor-positive/HER2-negative breast cancers. However, detailed studies comparing TROP2 protein expression in the different molecular subtypes of breast cancer are limited, and definitive evidence supporting the use of TROP2 as a biomarker for predicting response to this agent in patients with breast cancer is currently lacking. OBJECTIVE To compare the expression of TROP2 in the different molecular subtypes of breast cancer. METHODS Immunohistochemical staining for TROP2 was performed on 94 therapy-naive primary invasive breast carcinomas, including 25 luminal A-like, 25 luminal B-like, 19 HER2-like, and 25 triple-negative tumors. RESULTS Intermediate to high levels of TROP2 expression were observed in the majority of carcinomas of each molecular subtype, with a wide range of expression in each subtype. Occasional tumors with low or absent TROP2 expression were encountered, including two metaplastic carcinomas which were completely negative for TROP2. CONCLUSIONS Our observations support the continued investigation of the efficacy of sacituzumab govitecan in all molecular subtypes of breast carcinoma. Furthermore, the observed wide range of expression of TROP2 suggests that TROP2 may have potential utility as a biomarker for predicting responsiveness to sacituzumab govitecan. If this proves to be the case, then immunohistochemical staining for TROP2 would be critical for identifying those patients whose tumors are completely negative for TROP2, since these patients may be least likely or unlikely to respond to this agent, and alternative therapies may be more appropriate in such instances.
Collapse
Affiliation(s)
- Richard B Mertens
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA.
| | - Elias P Makhoul
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Xiaomo Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Farnaz Dadmanesh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| |
Collapse
|
28
|
Keskinkılıc M, Gökmen-Polar Y, Badve SS. Triple Negative Breast Cancers: An Obsolete Entity? Clin Breast Cancer 2024; 24:1-6. [PMID: 38016912 DOI: 10.1016/j.clbc.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 11/30/2023]
Abstract
Triple negative breast cancer is defined on the basis of what it is not. It has served as a useful umbrella entity for management of patients with breast cancer for the last couple of decades. However, during this period a number of novel therapies have become available. These therapies have been documented to be useful in subsets of TNBCs that can be identified on the basis of distinct biologic alterations. Herein we revisit the categorization and usage of the TNBC as an entity to assess its utility in view of the currently available therapies.
Collapse
Affiliation(s)
- Merve Keskinkılıc
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Yesim Gökmen-Polar
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Sunil S Badve
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA.
| |
Collapse
|
29
|
Zhuang W, Zhang W, Xie L, Wang L, Li Y, Wang Z, Zhang A, Qiu H, Feng J, Zhang B, Hu Y. Generation and Characterization of SORT1-Targeted Antibody-Drug Conjugate for the Treatment of SORT1-Positive Breast Tumor. Int J Mol Sci 2023; 24:17631. [PMID: 38139459 PMCID: PMC10743877 DOI: 10.3390/ijms242417631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Antibody-drug conjugates (ADCs) have greatly improved the outcomes of advanced breast tumors. However, the treatment of breast tumors with existing ADCs is still hindered by many issues, such as tumor antigen heterogeneity and drug resistance. Therefore, ADCs against new targets would provide options for the treatment of these challenges. Sortilin-1 (SORT1) may be a promising target for ADC as it is upregulated in breast cancer. To evaluate the possibility of SORT1 as an ADC target, a humanized antibody_8D302 with high affinity against SORT1 was generated. Additionally, 8D302 was conjugated with MMAE and DXd to generate two ADCs_8D302-MMAE and 8D302-DXd, respectively. Both 8D302-MMAE and 8D302-DXd showed effective cytotoxicity against SORT1 positive breast tumor cell lines and induced bystander killing. Consequently, 8D302-MMAE showed relatively better anti-tumor activity than 8D302-DXd both in vitro and in vivo, but 8D302-DXd had superior safety profile and pharmacokinetics profile over 8D302-MMAE. Furthermore, SORT1 induced faster internalization and lysosomal trafficking of antibodies and had a higher turnover compared with HER2. Also, 8D302-DXd exhibited superior cell cytotoxicity and tumor suppression over trastuzumab-DXd, a HER2-targeted ADC. We hypothesize that the high turnover of SORT1 enables SORT1-targeted ADC to be a powerful agent for the treatment of SORT1-positive breast tumor.
Collapse
Affiliation(s)
- Weiliang Zhuang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; (W.Z.); (L.W.)
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Wei Zhang
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Liping Xie
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Lei Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; (W.Z.); (L.W.)
| | - Yuan Li
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Ziyu Wang
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Ao Zhang
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Haitao Qiu
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Jun Feng
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Baohong Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; (W.Z.); (L.W.)
| | - Youjia Hu
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| |
Collapse
|
30
|
Kalinkin AI, Sigin VO, Kuznetsova EB, Ignatova EO, Vinogradov II, Vinogradov MI, Vinogradov IY, Zaletaev DV, Nemtsova MV, Kutsev SI, Tanas AS, Strelnikov VV. Epigenomic Profiling Advises Therapeutic Potential of Leukotriene Receptor Inhibitors for a Subset of Triple-Negative Breast Tumors. Int J Mol Sci 2023; 24:17343. [PMID: 38139172 PMCID: PMC10743620 DOI: 10.3390/ijms242417343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive molecular subtype, with a poor survival rate compared to others subtypes. For a long time, chemotherapy was the only systemic treatment for TNBC, and the identification of actionable molecular targets might ultimately improve the prognosis for TNBC patients. We performed a genome-wide analysis of DNA methylation at CpG islands on a collection of one hundred ten breast carcinoma samples and six normal breast tissue samples using reduced representation bisulfite sequencing with the XmaI restriction enzyme (XmaI-RRBS) and identified a subset of TNBC samples with significant hypomethylation at the LTB4R/LTB4R2 genes' CpG islands, including CpG dinucleotides covered with cg12853742 and cg21886367 HumanMethylation 450K microarray probes. Abnormal DNA hypomethylation of this region in TNBC compared to normal samples was confirmed by bisulfite Sanger sequencing. Gene expression generally anticorrelates with promoter methylation, and thus, the promoter hypomethylation detected and confirmed in our study might be revealed as an indirect marker of high LTB4R/LTB4R2 expression using a simple methylation-sensitive PCR test. Analysis of RNA-seq expression and DNA methylation data from the TCGA dataset demonstrates that the expression of the LTB4R and LTB4R2 genes significantly negatively correlates with DNA methylation at both CpG sites cg12853742 (R = -0.4, p = 2.6 × 10-6; R = -0.21, p = 0.015) and cg21886367 (R = -0.45, p = 7.3 × 10-8; R = -0.24, p = 0.005), suggesting the upregulation of these genes in tumors with abnormal hypomethylation of their CpG island. Kaplan-Meier analysis using the TCGA-BRCA gene expression and clinical data revealed poorer overall survival for TNBC patients with an upregulated LTB4R. To this day, only the leukotriene inhibitor LY255283 has been tested on an MCF-7/DOX cell line, which is a luminal A breast cancer molecular subtype. Other studies compare the effects of Montelukast and Zafirlukast (inhibitors of the cysteinyl leukotriene receptor, which is different from LTB4R/LTB4R2) on the MDA-MB-231 (TNBC) cell line, with high methylation and low expression levels of LTB4R. In our study, we assess the therapeutic effects of various drugs (including leukotriene receptor inhibitors) with the DepMap gene effect and drug sensitivity data for TNBC cell lines with hypomethylated and upregulated LTB4R/LTB4R2 genes. LY255283, Minocycline, Silibinin, Piceatannol, Mitiglinide, 1-Azakenpaullone, Carbetocin, and Pim-1-inhibitor-2 can be considered as candidates for the additional treatment of TNBC patients with tumors demonstrating LTB4R/LTB4R2 hypomethylation/upregulation. Finally, our results suggest that the epigenetic status of leukotriene B4 receptors is a novel, potential, predictive, and prognostic biomarker for TNBC. These findings might improve individualized therapy for TNBC patients by introducing new therapeutic adjuncts as anticancer agents.
Collapse
Affiliation(s)
- Alexey I. Kalinkin
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
| | - Vladimir O. Sigin
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
| | - Ekaterina B. Kuznetsova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
- Laboratory of Medical Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119992 Moscow, Russia
| | - Ekaterina O. Ignatova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
- Nikolay Nikolaevich Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
| | - Ilya I. Vinogradov
- Regional Clinical Oncology Dispensary, 390011 Ryazan, Russia;
- Department of Histology, Pathological Anatomy and Medical Genetics, Ryazan State Medical University, 390026 Ryazan, Russia; (M.I.V.); (I.Y.V.)
| | - Maxim I. Vinogradov
- Department of Histology, Pathological Anatomy and Medical Genetics, Ryazan State Medical University, 390026 Ryazan, Russia; (M.I.V.); (I.Y.V.)
| | - Igor Y. Vinogradov
- Department of Histology, Pathological Anatomy and Medical Genetics, Ryazan State Medical University, 390026 Ryazan, Russia; (M.I.V.); (I.Y.V.)
| | - Dmitry V. Zaletaev
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
| | - Marina V. Nemtsova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
- Laboratory of Medical Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119992 Moscow, Russia
| | - Sergey I. Kutsev
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
| | - Alexander S. Tanas
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
| | - Vladimir V. Strelnikov
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (V.O.S.); (E.B.K.); (E.O.I.); (D.V.Z.); (M.V.N.); (S.I.K.); (A.S.T.); (V.V.S.)
| |
Collapse
|
31
|
Thomas A, Douglas E, Reis-Filho JS, Gurcan MN, Wen HY. Metaplastic Breast Cancer: Current Understanding and Future Directions. Clin Breast Cancer 2023; 23:775-783. [PMID: 37179225 PMCID: PMC10584986 DOI: 10.1016/j.clbc.2023.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/16/2023] [Indexed: 05/15/2023]
Abstract
Metaplastic breast cancers (MBC) encompass a group of highly heterogeneous tumors which share the ability to differentiate into squamous, mesenchymal or neuroectodermal components. While often termed rare breast tumors, given the relatively high prevalence of breast cancer, they are seen with some frequency. Depending upon the definition applied, MBC represents 0.2% to 1% of breast cancers diagnosed in the United States. Less is known about the epidemiology of MBC globally, though a growing number of reports are providing information on this. These tumors are often more advanced at presentation relative to breast cancer broadly. While more indolent subtypes exist, the majority of MBC subtypes are associated with inferior survival. MBC is most commonly of triple-negative phenotype. In less common hormone receptor positive MBCs, hormone receptor status appears not to be prognostic. In contrast, relatively rare HER2-positive MBCs are associated with superior outcomes. Multiple potentially targetable molecular features are overrepresented in MBC including DNA repair deficiency signatures and PIK3/AKT/mTOR and WNT pathways alterations. Data on the prevalence of targets for novel antibody-drug conjugates is also emerging. While chemotherapy appears to be less active in MBC than in other breast cancer subtypes, efficacy is seen in some MBCs. Disease-specific trials, as well as reports of exceptional responses, may provide clues for novel approaches to this often hard-to-treat breast cancer. Strategies which harness newer research tools, such as large data and artificial intelligence hold the promise of overcoming historic barriers to the study of uncommon tumors and could markedly advance disease-specific understanding in MBC.
Collapse
Affiliation(s)
- Alexandra Thomas
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC.
| | - Emily Douglas
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Metin N Gurcan
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Hannah Y Wen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
32
|
Yao L, Chen J, Ma W. Decoding TROP2 in breast cancer: significance, clinical implications, and therapeutic advancements. Front Oncol 2023; 13:1292211. [PMID: 37954074 PMCID: PMC10635515 DOI: 10.3389/fonc.2023.1292211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
Breast cancer is a heterogeneous disease characterized by distinct molecular subtypes, varied prognoses, and differential treatment responses. Understanding the molecular landscape and identifying therapeutic targets, such as trophoblast cell-surface antigen 2 (TROP2), is vital. TROP2 is notably overexpressed in breast cancer, playing a significant role in tumor growth, invasion, metastasis, and treatment resistance. While significant progress has been made in targeting TROP2 in breast cancer, several challenges and knowledge gaps remain. These challenges include the heterogeneity of TROP2 expression within breast cancer subtypes, resistance to its targeted therapies, potential off-target effects, limited therapeutic agents, and identifying optimal combination treatments. Integrating findings from clinical trials into clinical practice further complicates the landscape. This review article delves deep into TROP2 in breast cancer, highlighting its expression patterns, clinical implications, and therapeutic advancements. By understanding the role of TROP2, we can pave the way for personalized treatments, and transform the landscape of breast cancer care.
Collapse
Affiliation(s)
- Liqin Yao
- Department of Breast Surgical Oncology, The First Affiliated Hospital, Huzhou University School of Medicine, Huzhou, Zhejiang, China
| | - Junfeng Chen
- Department of Pathology and Clinical Laboratories, Tongxu County Hospital of Traditional Chinese Medicine, Kaifeng, Henan, China
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center, and Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
33
|
Zhang D, Lin J, Xu Y, Wu X, Xu X, Xie Y, Pan T, He Y, Luo J, Zhang Z, Fan L, Li S, Chen T, Wu A, Shao G. A novel dual-function SERS-based identification strategy for preliminary screening and accurate diagnosis of circulating tumor cells. J Mater Chem B 2023; 11:9666-9675. [PMID: 37779509 DOI: 10.1039/d3tb01545a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Non-specific adsorption of bioprobes based on surface-enhanced Raman spectroscopy (SERS) technology inevitably endows white blood cells (WBC) in the peripheral blood with Raman signals, which greatly interfere the identification accuracy of circulating tumor cells (CTCs). In this study, an innovative strategy was proposed to effectively identify CTCs by using SERS technology assisted by a receiver operating characteristic (ROC) curve. Firstly, a magnetic Fe3O4-Au complex SERS bioprobe was developed, which could effectively capture the triple negative breast cancer (TNBC) cells and endow the tumor cells with distinct SERS signals. Then, the ROC curve obtained based on the comparison of SERS intensity of TNBC cells and WBC was used to construct a tumor cell identification model. The merit of the model was that the detection sensitivity and specificity could be intelligently switched according to different identification purposes such as accurate diagnosis or preliminary screening of tumor cells. Finally, the difunctional recognition ability of the model for accurate diagnosis and preliminary screening of tumor cells was further validated by using the healthy human blood added with TNBC cells and blood samples of real tumor patients. This novel difunctional identification strategy provides a new perspective for identification of CTCs based on the SERS technology.
Collapse
Affiliation(s)
- Dinghu Zhang
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Jie Lin
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Yanping Xu
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Xiaoxia Wu
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Xiawei Xu
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Yujiao Xie
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Ting Pan
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Yiwei He
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Jun Luo
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Zhewei Zhang
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - LinYin Fan
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Shunxiang Li
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Tianxiang Chen
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Aiguo Wu
- Ningbo Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, P. R. China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Guoliang Shao
- Department of Interventional Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
34
|
D’Arienzo A, Verrazzo A, Pagliuca M, Napolitano F, Parola S, Viggiani M, Caputo R, Puglisi F, Giuliano M, Del Mastro L, Arpino G, De Laurentiis M, Montemurro F. Toxicity profile of antibody-drug conjugates in breast cancer: practical considerations. EClinicalMedicine 2023; 62:102113. [PMID: 37554126 PMCID: PMC10404866 DOI: 10.1016/j.eclinm.2023.102113] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Antibody-drug conjugates (ADCs) represent a novel and evolving class of antineoplastic agents, constituted by monoclonal antibody linked to biologically active drugs, delivering cytotoxic compounds at the tumor site, reducing the likelihood of systemic exposure and toxicity. They are generally well tolerated, nevertheless some predictable adverse reactions need careful monitoring and timely approach. These include neutropenia, nausea and vomiting, alopecia, diarrhea, left ventricular dysfunction, ILD/pneumonitis. The mechanisms leading to drug-associated toxicities are summarized, and prophylaxis protocols and appropriate management strategies are proposed, based on current literature. This review aims to collect the most updated evidence on toxicities potentially occurring during breast cancer treatment with approved or under clinical investigation (advanced stage) ADCs. A focus is dedicated to monitoring protocols and clinical management, aimed at preventing and/or promptly address relevant problems, in order to avoid premature discontinuation or improper dose reduction.
Collapse
Affiliation(s)
- Andrea D’Arienzo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Annarita Verrazzo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Via Mezzocannone 4, Naples 80138, Italy
| | - Martina Pagliuca
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Via Mezzocannone 4, Naples 80138, Italy
- Molecular Predictors and New Targets in Oncology Unit 981, Gustave Roussy, 114 Rue Édouard-Vaillant, Villejuif 94805, France
| | - Fabiana Napolitano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Sara Parola
- Oncology Unit, PO di San Felice a Cancello, Via Roma 349, San Felice a Cancello, Caserta 81021, Italy
| | - Martina Viggiani
- Department of Oncology, HFR Fribourg-Cantonal Hospital, Chemin des Pensionnats 2-6, Fribourg 1708, Switzerland
| | - Roberta Caputo
- Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola 52, Naples 80131, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Via Palladio 8, Udine 33100, Italy
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Via Franco Gallini 2, Aviano, Pordenone 33081, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Lucia Del Mastro
- Department of Medical Oncology, UO Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, Genova 16132, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Michelino De Laurentiis
- Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola 52, Naples 80131, Italy
| | - Filippo Montemurro
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 -KM 3.95, Candiolo, Torino 10060, Italy
| |
Collapse
|
35
|
Nerone M, Rossi L, Condorelli R, Ratti V, Conforti F, Palazzo A, Graffeo R. Beyond PARP Inhibitors in Advanced Breast Cancer Patients with Germline BRCA1/2 Mutations: Focus on CDK4/6-Inhibitors and Data Review on Other Biological Therapies. Cancers (Basel) 2023; 15:3305. [PMID: 37444415 DOI: 10.3390/cancers15133305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
We explored the outcomes of germline BRCA1/2 pathogenic/likely pathogenic variants (PVs/LPVs) in the endocrine-sensitive disease treated with first-line standard of care cyclin-dependent kinase 4/6 (CDK4/6) inhibitors. Three studies retrospectively showed a reduction in the overall survival (OS) and progression-free survival (PFS) in gBRCA1/2m patients compared to both the germinal BRCA1/2 wild type (gBRCA1/2wt) and the untested population. Regarding the efficacy of PI3Kα inhibitors, there are no subgroups or biomarker analyses in which germinal BRCA status was explored. However, the biological interactions between the PIK3CA/AKT/mTOR pathway and BRCA1/2 at a molecular level could help us to understand the activity of these drugs when used to treat BC in BRCA1/2 PVs/LPVs carriers. The efficacy of trastuzumab deruxtecan (T-DXd), an antibody-drug conjugate (ADC) targeting HER2 for HER2-low and HER2-positive (HER2+) BC, has been increasingly described. Unfortunately, data on T-DXd in HER2+ or HER2-low metastatic BC harboring germinal BRCA1/2 PVs/LPVs is lacking. Including germinal BRCA1/2 status in the subgroup analysis of the registration trials of this ADC would be of great interest, especially in the phase III trial DESTINY-breast04. This trial enrolled patients with HER2-negative (HER2-) and both HR+ and HR- metastatic disease, which can now be categorized as HER2-low. The HER2-low subgroup includes tumors that were previously classified as triple negative, so it is highly likely that some women were germline BRCA1/2 PVs/LPVs carriers and this data was not reported. Germline BRCA1/2 status will be available for a higher number of individuals with BC in the near future, and data on the prognostic and predictive role of these PVs/LPVs is needed in order to choose the best treatment options.
Collapse
Affiliation(s)
- Marta Nerone
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| | - Lorenzo Rossi
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| | - Rosaria Condorelli
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| | - Vilma Ratti
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| | - Fabio Conforti
- Oncology Unit, Humanitas Gavazzeni, 24125 Bergamo, Italy
| | - Antonella Palazzo
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Rossella Graffeo
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| |
Collapse
|
36
|
Ivanova M, Porta FM, Giugliano F, Frascarelli C, Sajjadi E, Venetis K, Cursano G, Mazzarol G, Guerini-Rocco E, Curigliano G, Criscitiello C, Fusco N. Breast Cancer with Brain Metastasis: Molecular Insights and Clinical Management. Genes (Basel) 2023; 14:1160. [PMID: 37372340 DOI: 10.3390/genes14061160] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer is the most frequently diagnosed malignancy worldwide and the leading cause of cancer-related death among women. Brain metastases are a primary contributor to mortality, as they often go undetected until late stages due to their dormant nature. Moreover, the clinical management of brain metastases is complicated by the relevant issue of blood-brain barrier penetration. The molecular pathways involved in the formation, progression, and colonization of primary breast tumors and subsequent brain metastases are diverse, posing significant hurdles due to the heterogeneous nature of breast cancer subtypes. Despite advancements in primary breast cancer treatments, the prognosis for patients with brain metastases remains poor. In this review, we aim to highlight the biological mechanisms of breast cancer brain metastases by evaluating multi-step genetic pathways and to discuss currently available and emerging treatment strategies to propose a prospective overview of the management of this complex disease.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Francesca Maria Porta
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- School of Pathology, University of Milan, 20122 Milan, Italy
| | - Federica Giugliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giovanni Mazzarol
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
37
|
Li J, Goh ELK, He J, Li Y, Fan Z, Yu Z, Yuan P, Liu DX. Emerging Intrinsic Therapeutic Targets for Metastatic Breast Cancer. BIOLOGY 2023; 12:697. [PMID: 37237509 PMCID: PMC10215321 DOI: 10.3390/biology12050697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
Breast cancer is now the most common cancer worldwide, and it is also the main cause of cancer-related death in women. Survival rates for female breast cancer have significantly improved due to early diagnosis and better treatment. Nevertheless, for patients with advanced or metastatic breast cancer, the survival rate is still low, reflecting a need for the development of new therapies. Mechanistic insights into metastatic breast cancer have provided excellent opportunities for developing novel therapeutic strategies. Although high-throughput approaches have identified several therapeutic targets in metastatic disease, some subtypes such as triple-negative breast cancer do not yet have an apparent tumor-specific receptor or pathway to target. Therefore, exploring new druggable targets in metastatic disease is a high clinical priority. In this review, we summarize the emerging intrinsic therapeutic targets for metastatic breast cancer, including cyclin D-dependent kinases CDK4 and CDK6, the PI3K/AKT/mTOR pathway, the insulin/IGF1R pathway, the EGFR/HER family, the JAK/STAT pathway, poly(ADP-ribose) polymerases (PARP), TROP-2, Src kinases, histone modification enzymes, activated growth factor receptors, androgen receptors, breast cancer stem cells, matrix metalloproteinases, and immune checkpoint proteins. We also review the latest development in breast cancer immunotherapy. Drugs that target these molecules/pathways are either already FDA-approved or currently being tested in clinical trials.
Collapse
Affiliation(s)
- Jiawei Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Eyleen L. K. Goh
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Ji He
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Yan Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Zhimin Fan
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan 250033, China;
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dong-Xu Liu
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| |
Collapse
|