1
|
Aibara D, Matsusue K. Glycoprotein nonmetastatic melanoma protein B is regulated by hepatic peroxisome proliferator-activated receptor γ in liver steatosis. Cell Signal 2025; 130:111678. [PMID: 39971219 DOI: 10.1016/j.cellsig.2025.111678] [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: 12/12/2024] [Revised: 02/09/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Hepatic glycoprotein nonmetastatic melanoma protein B (GPNMB) and nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) play essential roles in lipid metabolism. This study aimed to examine the molecular mechanism through which PPARγ controls GPNMB expression in liver steatosis. A microarray database was used to examine the gene expression patterns associated with fatty liver in type 2 diabetic leptin-deficient (ob/ob) mice, as well as in patients with non-alcoholic fatty liver disease (NAFLD) and advanced NAFLD. GPNMB expression significantly increased in the fatty livers of humans and mice. Elevated Gpnmb expression was notably reduced by liver-specific Pparγ knockout (PPARγLKO) in ob/ob mice. Similarly, alcohol-fed mice had increased hepatic Gpnmb levels. Transcriptomic analysis of the human liver samples revealed that Gpnmb expression was markedly higher in patients with fatty liver diseases, including those with NAFLD and alcoholic fatty liver disease, than in controls. Reporter and electrophoretic mobility shift assays confirmed that PPARγ directly enhances Gpnmb transcription via three functional PPARγ-responsive elements within the first intron. In conclusion, these findings suggest that Gpnmb is a novel PPARγ target in liver steatosis.
Collapse
Affiliation(s)
- Daisuke Aibara
- Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Kimihiko Matsusue
- Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
2
|
Jin YY, Desai VS, Mazzaroth J, Wickstrom E. IGF1R-Targeted Delivery of a Bridged Nucleic Acid Oligonucleotide-Peptide Conjugate for MicroRNA-21 Inhibition in Triple-Negative Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642231. [PMID: 40161818 PMCID: PMC11952343 DOI: 10.1101/2025.03.09.642231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Triple-negative breast cancer (TNBC), defined by the absence of ER, PR, and Her2, impacts over 46,000 U.S. women annually, disproportionately affecting minority ethnic groups and individuals with BRCA1 mutations. Despite advancements such as PARP inhibitors, TNBC remains highly aggressive, with frequent recurrences and a 50% mortality rate within four years, underscoring the urgent need for more effective targeted therapies. MicroRNAs (miRNAs) represent a novel therapeutic approach. In TNBC, overexpressed miR-21 drives tumor progression, immune evasion, treatment resistance, and metastasis. Targeted miR-21 inhibition could curb these effects while minimizing harm to normal cells. We developed a peptide-conjugated miR-21 inhibitor targeting TNBC cells via the overexpressed IGF1 receptor (IGF1R), associated with poor prognosis. Using aminomethyl-bridged nucleic acid (BNA) chemistry, a serum-stable, low-toxicity anti-miR-21 RNA analog was created and tested for its effects on TNBC cell proliferation, apoptosis, tumor suppressor expression, and immune checkpoint regulation. Conjugation to an IGF1 peptide analog improved delivery, demonstrating tumor-specific biodistribution, efficacy, and safety in TNBC-bearing mice. The miR-21 inhibitor-peptide conjugate reduced proliferation, induced apoptosis, elevated tumor suppressors, and suppressed immune checkpoints in TNBC cell lines. In vivo , it targeted tumors, halted growth, and showed no liver or kidney toxicity, supporting its potential as a targeted, low-toxicity TNBC therapy.
Collapse
|
3
|
Qiu Y, Shi Y, Chao Z, Zhu X, Chen Y, Lu L. Recent advances of antibody-drug conjugates in treating breast cancer with different HER2 status. Ther Adv Med Oncol 2025; 17:17588359241311379. [PMID: 39759831 PMCID: PMC11700426 DOI: 10.1177/17588359241311379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025] Open
Abstract
Despite the availability of multiple treatment options for breast cancer, challenges such as adverse events, drug resistance, and disease progression persist for patients. The identification of human epidermal growth factor receptor 2 (HER2) as an oncogenic driver in a subset of breast cancers, alongside the development of HER2-targeted therapies, has significantly improved the prognosis of HER2-amplified breast cancers. However, therapeutic options remain limited for HER2-overexpressing or HER2-negative breast cancers. In response to this gap, antibody-drug conjugates (ADCs) have emerged as a promising approach. ADCs combine the specificity of monoclonal antibodies with the cytotoxic effects of chemotherapy, which allows for the targeted delivery of a cytotoxic payload to cancer cells. ADCs have been used as adjuvant chemotherapeutic treatments and salvage therapies across various breast cancer subtypes, which have greatly improved the prognosis of breast cancer patients. Numerous ongoing clinical trials seek to optimize dosing strategies and identify patient populations that would benefit most from ADCs. This review presents an updated and comprehensive overview of emerging investigational ADCs for treating breast cancer patients with various HER2 subtypes. These ADCs are spearheading a new era in targeted cancer therapy, promising to innovate treatment paradigms for both HER2-positive and HER2-low breast cancers.
Collapse
Affiliation(s)
- Yue Qiu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yaqin Shi
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhujun Chao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xinyu Zhu
- Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yan Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Linlin Lu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| |
Collapse
|
4
|
Timis T, Buruiana S, Dima D, Nistor M, Muresan XM, Cenariu D, Tigu AB, Tomuleasa C. Advances in Cell and Immune Therapies for Melanoma. Biomedicines 2025; 13:98. [PMID: 39857682 PMCID: PMC11761552 DOI: 10.3390/biomedicines13010098] [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: 11/28/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
The incidence rate of cutaneous melanoma is on the rise worldwide, due to increased exposure to UV radiation, aging populations, and exposure to teratogen agents. However, diagnosis is more precise, and the increased number of new cases is related to the improved diagnosis tools. Despite better early diagnosis and better therapies, melanoma has remained a significant public health challenge because of its aggressive behavior and high potential for metastasis. In 2020, cutaneous melanoma constituted approximately 1.3% of all cancer deaths that occurred within the European Union, thereby highlighting the necessity for effective prevention, timely diagnosis, and sustainable treatment measures, especially as a growing number of cases occur among younger patients. Melanoma is regarded as one of the most inflamed cancers due to its high immune cell presence and strong response to immunotherapy, fueling the need for development of immune-driven innovative treatments. Approved therapies, including immune checkpoint inhibitors (e.g., anti-PD-1 and anti-CTLA-4), have notably improved survival rates in melanoma. However, the limitations of the PD-1/PD-L1 and CTLA-4 axes inhibitors, such as low response rates, treatment resistance, and toxicity, have driven the need for continued research and advancements in treatment strategies. Current clinical trials are exploring various combinations of immune checkpoint inhibitors with costimulatory receptor agonists, chemotherapy, targeted therapies, and other immunotherapies, with the goal of improving outcomes and reducing side effects for melanoma patients. Emerging approaches, including adoptive cell therapy with tumor-infiltrating lymphocytes (TILs) and oncolytic virotherapy, are showing promise. While CAR-T cell therapy has been less successful in melanoma compared to blood cancers, ongoing research is addressing challenges like the tumor microenvironment and antigen specificity. This review provides an overview of the requirement for advances in these medications, to mark a significant step forward in melanoma management, set to bring a fresh breath of hope for patients.
Collapse
Affiliation(s)
- Tanase Timis
- Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Department of Oncology, Bistrița Emergency Hospital, 420094 Bistrița, Romania
| | - Sanda Buruiana
- Department of Hematology, Nicolae Testemițanu University of Medicine and Pharmacy, MD-2004 Chisinau, Moldova
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Madalina Nistor
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ximena Maria Muresan
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Diana Cenariu
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Adrian-Bogdan Tigu
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
5
|
Sharma N, Bhati A, Aggarwal S, Shah K, Dewangan HK. PARP Pioneers: Using BRCA1/2 Mutation-targeted Inhibition to Revolutionize Breast Cancer Treatment. Curr Pharm Des 2025; 31:663-673. [PMID: 39421986 DOI: 10.2174/0113816128322894241004051814] [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: 05/06/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Breast cancer stands on the second position in the world in being common and women happen to have it with high rate of about five-folds around the world. The causes of occurrence can matter with different humans be it external factors or the internal genetic ones. Breast cancer is primarily driven by mutations in the BRCA1 and BRCA2 susceptibility genes. These BC susceptibility genes encode proteins critical for DNA homologous recombination repair (HRR). Poly (ADP ribose) polymerases (PARP) are the essential enzymes involved in the repairing of the damaged DNA. So the inhibition of these inhibitors can be considered as the promising strategy for targeting cancers with defective damage in the deoxyribonucleic acid. Olaparib and talazoparib are PARP inhibitors (PARPi) are being employed for the monotherapies in case of the deleterious germline HER2-negative and BRCA-mutated breast cancer. The potency of PARP for trapping on DNA and causes cytotoxicity may have difference in the safety and efficacy with the PARPi. The PARPi have been found its place in the all different types of breast cancers and have shown potential benefits. The purpose of this review is to provide an update on the oral poly (ADP-ribose) polymerase (PARP) inhibitors for the improvement in the treatment and management of breast cancer.
Collapse
Affiliation(s)
- Navneet Sharma
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Akash Bhati
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Shagun Aggarwal
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Kamal Shah
- Institute of Pharmaceutical Research (IPR), GLA University, NH-2, Delhi Mathura Road, PO-Chaumuhan, Mathura, Uttar Pradesh, India
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| |
Collapse
|
6
|
Belloni S, Tiberio P, De Sanctis R, Magon A, Santoro A, Zambelli A, Caruso R, Arrigoni C. Prevalence of treatment-related adverse events (TRAEs) with antibody-drug conjugates in metastatic breast cancer patients: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2024; 204:104527. [PMID: 39395623 DOI: 10.1016/j.critrevonc.2024.104527] [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: 07/12/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/14/2024] Open
Abstract
Antibody-drug conjugates (ADCs) are revolutionizing metastatic breast cancer treatment, resulting in a better prognosis and a higher safety profile than chemotherapy. Nevertheless, treatment-related adverse events (TRAE) have been extensively documented. We searched five databases for articles published up to December 2023 and conducted a meta-analysis on 23 clinical trials to estimate TRAE prevalence related to currently approved ADCs. The prevalence of the most common TRAEs ranged from 12 % to 33 %, depending on the ADC type and study design. Gastrointestinal disorders were highly prevalent during Trastuzumab Deruxtecan, general disorders were extremely common during Trastuzumab Emtansine, and blood system disorders and gastrointestinal disorders were the most prevalent during Sacituzumab Govitecan. This study provides an estimate of ADC-related TRAEs for each treatment based on study design. Despite each ADC having specific toxicities, gastrointestinal symptoms were highly prevalent in all treatments. This study lays the groundwork for developing personalized risk-stratified care pathways.
Collapse
Affiliation(s)
- Silvia Belloni
- Department of Public Health, Experimental and Forensic Medicine, Section of Hygiene, University of Pavia, Pavia 27100, Italy
| | - Paola Tiberio
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI) 20072, Italy; Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI) 20089, Italy
| | - Rita De Sanctis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI) 20072, Italy; Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI) 20089, Italy.
| | - Arianna Magon
- Health Professions Research and Development Unit, IRCCS Policlinico San Donato, San Donato Milanese, (MI) 20097, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI) 20072, Italy; Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI) 20089, Italy
| | - Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI) 20072, Italy; Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI) 20089, Italy
| | - Rosario Caruso
- Health Professions Research and Development Unit, IRCCS Policlinico San Donato, San Donato Milanese, (MI) 20097, Italy; Department of Biomedical Sciences for Health, University of Milan, 20122 Milan, Italy
| | - Cristina Arrigoni
- Department of Public Health, Experimental and Forensic Medicine, Section of Hygiene, University of Pavia, Pavia 27100, Italy
| |
Collapse
|
7
|
Mukherjee A, Bandyopadhyay D. Targeted Therapy in Breast Cancer: Advantages and Advancements of Antibody-Drug Conjugates, a Type of Chemo-Biologic Hybrid Drugs. Cancers (Basel) 2024; 16:3517. [PMID: 39456611 PMCID: PMC11505910 DOI: 10.3390/cancers16203517] [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: 08/25/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Cancer is a significant health challenge globally, with millions of people affected every year, resulting in high morbidity and mortality. Although other treatment options are available with limitations, chemotherapy, either standalone or combined with other therapeutic procedures, is the most commonly used practice of treating cancer. In chemotherapy, cancer cells/malignant tumors are targeted; however, due to less target specificity, along with malignant cells, normal cells are also affected, which leads to various off-target effects (side effects) that impact the patient quality of life. Out of all the different types of cancers, breast cancer is the most common type of cancer in humans worldwide. Current anticancer drug discovery research aims to develop therapeutics with higher potency and lower toxicity, which is only possible through target-specific therapy. Antibody-drug conjugates (ADCs) are explicitly designed to target malignant tumors and minimize off-target effects by reducing systemic cytotoxicity. Several ADCs have been approved for clinical use and have shown moderate to good efficacy so far. Considering various aspects, chemotherapy and ADCs are useful in treating cancer. However, ADCs provide a more focused and less toxic approach, which is especially helpful in cases where resistance to chemotherapy (drug resistance) occurs and in the type of malignancies in which specific antigens are overexpressed. Ongoing ADC research aims to develop more target-specific cancer treatments. In short, this study presents a concise overview of ADCs specific to breast cancer treatment. This study provides insight into the classifications, mechanisms of action, structural aspects, and clinical trial phases (current status) of these chemo-biologic drugs (ADCs).
Collapse
Affiliation(s)
- Attrayo Mukherjee
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Patia, Bhubaneswar 751024, Odisha, India;
| | - Debasish Bandyopadhyay
- School of Integrative Biological and Chemical Sciences (SIBCS), University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- School of Earth, Environmental, and Marine Sciences (SEEMS), University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
8
|
Tuo X, Chen J, Hao C, Dai X, Zhu J, Tian S, Zhang Y, Wang F. Identification of GPNMB in endometrial cancer based on pan-cancer analysis and in vitro validation. Discov Oncol 2024; 15:489. [PMID: 39331202 PMCID: PMC11436576 DOI: 10.1007/s12672-024-01382-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND GPNMB is a type I transmembrane protein, and emerging evidence supports the relationship between GPNMB and cancers. OBJECTIVE Through a comprehensive pan-cancer analysis, we examined the expression levels, prognostic significance, and mutation profiles of GPNMB in different cancer types. Subsequently, utilizing in vitro experiments, we elucidated the impact of GPNMB in endometrial cancer (EC). METHODS TIMER2, GEPIA2, UALCAN and cBioPortal were used to analyze the expression pattern, prognostic values, and mutation status of GPNMB. HEC-1B and Ishikawa cells were used to conduct in vitro analyses of GPNMB overexpression. GeneMANIA and TIMER2 were used to evaluate the potential functions and correlations between GPNMB expression and tumor-infiltrating immune cells in EC. RESULTS GPNMB was found to be highly expressed in multiple cancers, where it was associated with poor prognosis. Additionally, GPNMB was downregulated at both mRNA and protein levels in EC. Overexpression of GPNMB inhibited the proliferation, migration, and invasion of HEC-1B and Ishikawa cells. Functional analysis showed that GPNMB was enriched in pathways associated with regulation of plasma lipoprotein particle levels. The expression of GPNMB was positively connected with B cell, CD8+ T cell, CD4+ T cell, Macrophage, Neutrophil, and Dendritic cell levels. CONCLUSION Through pan-cancer analysis, we identified the antitumor effect of GPNMB in EC and predicted the potential mechanisms between GPNMB expression and EC.
Collapse
Affiliation(s)
- Xiaoqian Tuo
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Jialan Chen
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Cuipei Hao
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Xiaole Dai
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Jiayi Zhu
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Siqi Tian
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Yan Zhang
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Fan Wang
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China.
| |
Collapse
|
9
|
Chang HP, Cheung YK, Liu S, Shah DK. Development of a generalized pharmacokinetic model to characterize clinical pharmacokinetics of monomethyl auristatin E-based antibody-drug conjugates. Br J Clin Pharmacol 2024; 90:1637-1655. [PMID: 38566392 DOI: 10.1111/bcp.16057] [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] [Received: 07/27/2023] [Revised: 01/30/2024] [Accepted: 02/10/2024] [Indexed: 04/04/2024] Open
Abstract
AIMS This study aims to develop a generalized pharmacokinetic (PK) model for monomethyl auristatin E (MMAE)-based antibody-drug conjugates (ADCs) that can simultaneously capture the PK of multiple ADC analytes commonly measured in the clinic. METHODS A comprehensive literature review was conducted to collect PK data on MMAE-based ADCs from clinical trials. From each study, PK profiles of total antibody, the ADC, conjugated MMAE, and unconjugated MMAE, were extracted. These data were pooled and dose-normalized to evaluate the generalizability of PK across various ADCs and dose levels. Upon confirming PK generalizability, a generalized PK model for MMAE-based ADCs was developed using the entire dataset. Furthermore, exposure metrics ( C max and AUC) reported across the range of doses were combined to establish linear relationships between dose and exposure metrics for MMAE-based ADCs. RESULTS A total of 109 PK profiles from 18 distinct MMAE-based ADCs were gathered. The dose-normalized PK profiles supported the generalizability of PK for MMAE-based ADCs. A generalized PK model was developed, which enabled capturing the PK data for 4 ADC analytes across all collected MMAE-based ADCs. A linear relationship between dose and PK exposure metrics was established, enabling the prediction of typical exposure values across different doses for MMAE-based ADCs. CONCLUSIONS This study comprehensively analysed clinical PK data from different valine-citrulline (vc)-MMAE-based ADCs. The generalized PK model developed here serves as an important tool for a priori prediction of the PK for multiple ADC analytes in clinical settings and lays the foundation for establishing generalized exposure-response and exposure-toxicity correlations for MMAE-based ADCs.
Collapse
Affiliation(s)
- Hsuan-Ping Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Yuen Kiu Cheung
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Shufang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
10
|
Bamodu OA, Chung CC, Pisanic TR, Wu ATH. The intricate interplay between cancer stem cells and cell-of-origin of cancer: implications for therapeutic strategies. Front Oncol 2024; 14:1404628. [PMID: 38800385 PMCID: PMC11116576 DOI: 10.3389/fonc.2024.1404628] [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: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Background Cancer stem cells (CSCs) have emerged as pivotal players in tumorigenesis, disease progression, and resistance to therapies. Objective This comprehensive review delves into the intricate relationship between CSCs and the cell-of-origin in diverse cancer types. Design Comprehensive review of thematically-relevant literature. Methods We explore the underlying molecular mechanisms that drive the conversion of normal cells into CSCs and the impact of the cell-of-origin on CSC properties, tumor initiation, and therapeutic responses. Moreover, we discuss potential therapeutic interventions targeting CSCs based on their distinct cell-of-origin characteristics. Results Accruing evidence suggest that the cell-of-origin, the cell type from which the tumor originates, plays a crucial role in determining the properties of CSCs and their contribution to tumor heterogeneity. Conclusion By providing critical insights into the complex interplay between CSCs and their cellular origins, this article aims to enhance our understanding of cancer biology and pave the way for more effective and personalized cancer treatments.
Collapse
Affiliation(s)
- Oluwaseun Adebayo Bamodu
- Directorate of Postgraduate Studies, School of Clinical Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Chen-Chih Chung
- Department of Neurology, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan
| | - Thomas R. Pisanic
- Johns Hopkins Institute for NanoBioTechnology, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology - Cancer Genetics and Epigenetics, Johns Hopkins University, Baltimore, MD, United States
| | - Alexander T. H. Wu
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
11
|
Kundu M, Butti R, Panda VK, Malhotra D, Das S, Mitra T, Kapse P, Gosavi SW, Kundu GC. Modulation of the tumor microenvironment and mechanism of immunotherapy-based drug resistance in breast cancer. Mol Cancer 2024; 23:92. [PMID: 38715072 PMCID: PMC11075356 DOI: 10.1186/s12943-024-01990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 04/02/2024] [Indexed: 05/12/2024] Open
Abstract
Breast cancer, the most frequent female malignancy, is often curable when detected at an early stage. The treatment of metastatic breast cancer is more challenging and may be unresponsive to conventional therapy. Immunotherapy is crucial for treating metastatic breast cancer, but its resistance is a major limitation. The tumor microenvironment (TME) is vital in modulating the immunotherapy response. Various tumor microenvironmental components, such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), are involved in TME modulation to cause immunotherapy resistance. This review highlights the role of stromal cells in modulating the breast tumor microenvironment, including the involvement of CAF-TAM interaction, alteration of tumor metabolism leading to immunotherapy failure, and other latest strategies, including high throughput genomic screening, single-cell and spatial omics techniques for identifying tumor immune genes regulating immunotherapy response. This review emphasizes the therapeutic approach to overcome breast cancer immune resistance through CAF reprogramming, modulation of TAM polarization, tumor metabolism, and genomic alterations.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
- Department of Pharmaceutical Technology, Brainware University, West Bengal, 700125, India
| | - Ramesh Butti
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Venketesh K Panda
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
| | - Sumit Das
- National Centre for Cell Sciences, Savitribai Phule Pune University Campus, Pune, 411007, India
| | - Tandrima Mitra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
| | - Prachi Kapse
- School of Basic Medical Sciences, Savitribai Phule Pune University, Pune, 411007, India
| | - Suresh W Gosavi
- School of Basic Medical Sciences, Savitribai Phule Pune University, Pune, 411007, India
| | - Gopal C Kundu
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India.
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Bhubaneswar, 751024, India.
| |
Collapse
|
12
|
Kimura T, Okita Y, Nagumo Y, Chin JM, Fikry MA, Shiga M, Kandori S, Kawahara T, Suzuki H, Nishiyama H, Kato M. Glycoprotein nonmetastatic melanoma protein B impacts the malignant potential of bladder cancer cells through its hem-immunoreceptor tyrosine-based activation motif. Pathol Int 2024; 74:262-273. [PMID: 38501371 DOI: 10.1111/pin.13419] [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] [Received: 12/25/2023] [Revised: 02/12/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
Bladder cancer is one of the most common cancers among men worldwide. Although multiple genomic mutations and epigenetic alterations have been identified, an efficacious molecularly targeted therapy has yet to be established. Therefore, a novel approach is anticipated. Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type I transmembrane glycoprotein that is highly expressed in various cancers. In this study, we evaluated bladder cancer patient samples and found that GPNMB protein abundance is associated with high-grade tumors, and both univariate and multivariate analyses showed that GPNMB is a prognostic factor. Furthermore, the prognosis of patients with high GPNMB levels was significantly poorer in those with nonmuscle invasive bladder cancer (NMIBC) than in those with muscle invasive bladder cancer (MIBC). We then demonstrated that knockdown of GPNMB in MIBC cell lines with high GPNMB inhibits cellular migration and invasion, whereas overexpression of GPNMB further enhances cellular migration and invasion in MIBC cell lines with originally low GPNMB. Therefore, we propose that GPNMB is one of multiple driver molecules in the acquisition of cellular migratory and invasive potential in bladder cancers. Moreover, we revealed that the tyrosine residue in the hemi-immunoreceptor tyrosine-based activation motif (hemITAM) is required for GPNMB-induced cellular motility.
Collapse
Affiliation(s)
- Tomokazu Kimura
- Department of Urology, University of Tsukuba, Ibaraki, Japan
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yukari Okita
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Division of Cell Dynamics, Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | | | - Jas Min Chin
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Muhammad Ali Fikry
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masanobu Shiga
- Department of Urology, University of Tsukuba, Ibaraki, Japan
| | - Shuya Kandori
- Department of Urology, University of Tsukuba, Ibaraki, Japan
| | | | - Hiroyuki Suzuki
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Mitsuyasu Kato
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Division of Cell Dynamics, Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
13
|
Filis P, Zerdes I, Soumala T, Matikas A, Foukakis T. The ever-expanding landscape of antibody-drug conjugates (ADCs) in solid tumors: A systematic review. Crit Rev Oncol Hematol 2023; 192:104189. [PMID: 37866413 DOI: 10.1016/j.critrevonc.2023.104189] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND The advent of targeted therapies signaled novel avenues for more optimal oncological outcomes. Antibody-drug conjugates (ADCs) have risen as a cornerstone of the ever-expanding targeted therapy era. The purpose of this systematic review is to delineate the rapidly evolving clinical landscape of ADCs for solid tumors. METHODS A literature search was performed in Medline, Embase and Cochrane databases for phase II and III clinical trials. Outcomes of interest were the objective response rate, overall survival, progression-free survival and adverse events. RESULTS A total of 92 clinical trials (76 phase II and 16 phase III) evaluated the efficacy and safety of ADCs for a plethora of solid tumors. Out of the 30 investigated ADCs, 8 have received approval by regulatory organizations for solid tumors. Currently, 52 phase III clinical trials for ADCs are ongoing. CONCLUSION ADCs have shown promising results for several solid tumors and various cancer settings.
Collapse
Affiliation(s)
- Panagiotis Filis
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece.
| | - Ioannis Zerdes
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Theodora Soumala
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Alexios Matikas
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
14
|
Püsküllüoğlu M, Rudzińska A, Pacholczak-Madej R. Antibody-drug conjugates in HER-2 negative breast cancers with poor prognosis. Biochim Biophys Acta Rev Cancer 2023; 1878:188991. [PMID: 37758021 DOI: 10.1016/j.bbcan.2023.188991] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
Antibody drug conjugates (ADCs) comprise a rapidly growing class of targeted drugs that selectively deliver a cytotoxic agent to cancer cells, reducing the side effects associated with conventional chemotherapy. Breast cancer (BC) is a heterogeneous entity. The need for effective therapies for HER-2 negative BCs with poor prognosis, such as triple-negative or endocrine-resistant BC, remains unmet due to the lack of potential targets for treatments. These BC subtypes are not candidates for hormonal or anti-HER-2 agents. However, ongoing clinical trials exploring the use of ADCs with a wide range of targets have shown potential for this treatment modality. In this review, we present the current state of knowledge regarding the role of ADC and speculate on novel approaches including ADC combination therapies, new molecular targets, and the role of other subclasses of ADCs (bicycle drug conjugates, bispecific ADCs, immune modulating ADCs) in this clinical scenario.
Collapse
Affiliation(s)
- Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Garncarska Street 11, 31-115 Krakow, Poland.
| | - Agnieszka Rudzińska
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Garncarska Street 11, 31-115 Krakow, Poland
| | - Renata Pacholczak-Madej
- Department of Anatomy, Jagiellonian University, Medical College, Kopernika Street 12, 31-034 Krakow, Poland; Department of Chemotherapy, The District Hospital, 22 Szpitalna Street, 34-200 Sucha Beskidzka, Poland
| |
Collapse
|
15
|
Tong L, Yu X, Wang S, Chen L, Wu Y. Research Progress on Molecular Subtyping and Modern Treatment of Triple-Negative Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:647-658. [PMID: 37644916 PMCID: PMC10461741 DOI: 10.2147/bctt.s426121] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Breast cancer has become the most common malignant tumor worldwide. Triple-negative breast cancer (TNBC) is a type of breast cancer that is negative for estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Compared with other molecular subtypes of breast cancer, TNBC is the most aggressive and highly heterogeneous. TNBC is insensitive to endocrine and anti-HER2 therapy, and chemotherapy is currently the main systemic treatment. With the continuous development of detection techniques and deepening research on TNBC molecular subtypes, drugs targeting immune checkpoints and different targets have emerged, such as atezolizumab, pembrolizumab, poly (ADP-ribose) polymerase (PARP) inhibitors, trophoblast cell-surface antigen 2 (TROP-2), and antibody-drug conjugates. These therapies provide new hope for TNBC treatment. Based on the analysis and classification of TNBC, this article summarizes the immunotherapy, targeted therapy, and new treatment combinations, providing references for the precise treatment of TNBC in the future.
Collapse
Affiliation(s)
- Ling Tong
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
- Department of Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| | - Xiangling Yu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| | - Shan Wang
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
- Department of Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| | - Ling Chen
- Department of Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| | - Yibo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| |
Collapse
|
16
|
Yang T, Li W, Huang T, Zhou J. Antibody-Drug Conjugates for Breast Cancer Treatment: Emerging Agents, Targets and Future Directions. Int J Mol Sci 2023; 24:11903. [PMID: 37569276 PMCID: PMC10418918 DOI: 10.3390/ijms241511903] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/21/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
To achieve the scheme of "magic bullets" in antitumor therapy, antibody-drug conjugates (ADCs) were developed. ADCs consist of antibodies targeting tumor-specific antigens, chemical linkers, and cytotoxic payloads that powerfully kill cancer cells. With the approval of ado-trastuzumab emtansine (T-DM1) and fam-trastuzumab deruxtecan (T-DXd), the therapeutic potentials of ADCs in breast cancer have come into the spotlight. Nearly 30 ADCs for breast cancer are under exploration to move targeted therapy forward. In this review, we summarize the presenting and emerging agents and targets of ADCs. The ADC structure and development history are also concluded. Moreover, the challenges faced and prospected future directions in this field are reviewed, which give insights into novel treatments with ADCs for breast cancer.
Collapse
Affiliation(s)
| | | | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
17
|
Lang M, Schmidt LS, Wilson KM, Ricketts CJ, Sourbier C, Vocke CD, Wei D, Crooks DR, Yang Y, Gibbs BK, Zhang X, Klumpp-Thomas C, Chen L, Guha R, Ferrer M, McKnight C, Itkin Z, Wangsa D, Wangsa D, James A, Difilippantonio S, Karim B, Morís F, Ried T, Merino MJ, Srinivasan R, Thomas CJ, Linehan WM. High-throughput and targeted drug screens identify pharmacological candidates against MiT-translocation renal cell carcinoma. J Exp Clin Cancer Res 2023; 42:99. [PMID: 37095531 PMCID: PMC10127337 DOI: 10.1186/s13046-023-02667-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/06/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND MiT-Renal Cell Carcinoma (RCC) is characterized by genomic translocations involving microphthalmia-associated transcription factor (MiT) family members TFE3, TFEB, or MITF. MiT-RCC represents a specific subtype of sporadic RCC that is predominantly seen in young patients and can present with heterogeneous histological features making diagnosis challenging. Moreover, the disease biology of this aggressive cancer is poorly understood and there is no accepted standard of care therapy for patients with advanced disease. Tumor-derived cell lines have been established from human TFE3-RCC providing useful models for preclinical studies. METHODS TFE3-RCC tumor derived cell lines and their tissues of origin were characterized by IHC and gene expression analyses. An unbiased high-throughput drug screen was performed to identify novel therapeutic agents for treatment of MiT-RCC. Potential therapeutic candidates were validated in in vitro and in vivo preclinical studies. Mechanistic assays were conducted to confirm the on-target effects of drugs. RESULTS The results of a high-throughput small molecule drug screen utilizing three TFE3-RCC tumor-derived cell lines identified five classes of agents with potential pharmacological efficacy, including inhibitors of phosphoinositide-3-kinase (PI3K) and mechanistic target of rapamycin (mTOR), and several additional agents, including the transcription inhibitor Mithramycin A. Upregulation of the cell surface marker GPNMB, a specific MiT transcriptional target, was confirmed in TFE3-RCC and evaluated as a therapeutic target using the GPNMB-targeted antibody-drug conjugate CDX-011. In vitro and in vivo preclinical studies demonstrated efficacy of the PI3K/mTOR inhibitor NVP-BGT226, Mithramycin A, and CDX-011 as potential therapeutic options for treating advanced MiT-RCC as single agents or in combination. CONCLUSIONS The results of the high-throughput drug screen and validation studies in TFE3-RCC tumor-derived cell lines have provided in vitro and in vivo preclinical data supporting the efficacy of the PI3K/mTOR inhibitor NVP-BGT226, the transcription inhibitor Mithramycin A, and GPNMB-targeted antibody-drug conjugate CDX-011 as potential therapeutic options for treating advanced MiT-RCC. The findings presented here should provide the basis for designing future clinical trials for patients with MiT-driven RCC.
Collapse
Affiliation(s)
- Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, 39100, Italy
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kelli M Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carole Sourbier
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cathy D Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Darmood Wei
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Youfeng Yang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin K Gibbs
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Carleen Klumpp-Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Lu Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Crystal McKnight
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Zina Itkin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - Darawalee Wangsa
- Genetics Branch, Cancer Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Danny Wangsa
- Genetics Branch, Cancer Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy James
- Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Simone Difilippantonio
- Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Baktir Karim
- Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Francisco Morís
- EntreChem SL, Vivero Ciencias de la Salud, Calle Colegio Santo Domingo Guzmán, Oviedo, AS, 33011, Spain
| | - Thomas Ried
- Genetics Branch, Cancer Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maria J Merino
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD, USA
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
18
|
Subhan MA, Parveen F, Shah H, Yalamarty SSK, Ataide JA, Torchilin VP. Recent Advances with Precision Medicine Treatment for Breast Cancer including Triple-Negative Sub-Type. Cancers (Basel) 2023; 15:2204. [PMID: 37190133 PMCID: PMC10137302 DOI: 10.3390/cancers15082204] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Breast cancer is a heterogeneous disease with different molecular subtypes. Breast cancer is the second leading cause of mortality in woman due to rapid metastasis and disease recurrence. Precision medicine remains an essential source to lower the off-target toxicities of chemotherapeutic agents and maximize the patient benefits. This is a crucial approach for a more effective treatment and prevention of disease. Precision-medicine methods are based on the selection of suitable biomarkers to envision the effectiveness of targeted therapy in a specific group of patients. Several druggable mutations have been identified in breast cancer patients. Current improvements in omics technologies have focused on more precise strategies for precision therapy. The development of next-generation sequencing technologies has raised hopes for precision-medicine treatment strategies in breast cancer (BC) and triple-negative breast cancer (TNBC). Targeted therapies utilizing immune checkpoint inhibitors (ICIs), epidermal growth factor receptor inhibitor (EGFRi), poly(ADP-ribose) polymerase inhibitor (PARPi), antibody-drug conjugates (ADCs), oncolytic viruses (OVs), glucose transporter-1 inhibitor (GLUT1i), and targeting signaling pathways are potential treatment approaches for BC and TNBC. This review emphasizes the recent progress made with the precision-medicine therapy of metastatic breast cancer and TNBC.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Farzana Parveen
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital Jhang 35200, Primary and Secondary Healthcare Department, Government of Punjab, Lahore 54000, Pakistan
| | - Hassan Shah
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Janaína Artem Ataide
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083-871, SP, Brazil
| | - Valdimir P. Torchilin
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
19
|
Nguyen TD, Bordeau BM, Balthasar JP. Mechanisms of ADC Toxicity and Strategies to Increase ADC Tolerability. Cancers (Basel) 2023; 15:713. [PMID: 36765668 PMCID: PMC9913659 DOI: 10.3390/cancers15030713] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Anti-cancer antibody-drug conjugates (ADCs) aim to expand the therapeutic index of traditional chemotherapy by employing the targeting specificity of monoclonal antibodies (mAbs) to increase the efficiency of the delivery of potent cytotoxic agents to malignant cells. In the past three years, the number of ADCs approved by the Food and Drug Administration (FDA) has tripled. Although several ADCs have demonstrated sufficient efficacy and safety to warrant FDA approval, the clinical use of all ADCs leads to substantial toxicity in treated patients, and many ADCs have failed during clinical development due to their unacceptable toxicity profiles. Analysis of the clinical data has demonstrated that dose-limiting toxicities (DLTs) are often shared by different ADCs that deliver the same cytotoxic payload, independent of the antigen that is targeted and/or the type of cancer that is treated. DLTs are commonly associated with cells and tissues that do not express the targeted antigen (i.e., off-target toxicity), and often limit ADC dosage to levels below those required for optimal anti-cancer effects. In this manuscript, we review the fundamental mechanisms contributing to ADC toxicity, we summarize common ADC treatment-related adverse events, and we discuss several approaches to mitigating ADC toxicity.
Collapse
Affiliation(s)
| | | | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
20
|
Al Jarroudi O, El Bairi K, Curigliano G, Afqir S. Antibody-Drug Conjugates: A New Therapeutic Approach for Triple-Negative Breast Cancer. Cancer Treat Res 2023; 188:1-27. [PMID: 38175340 DOI: 10.1007/978-3-031-33602-7_1] [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] [Indexed: 01/05/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subset associated with a worse prognosis and poor response to conventional chemotherapy. Despite recent advances in drug discovery, its management is still a challenge for clinicians, illuminating the unmet need to develop novel treatment approaches. Antibody-drug conjugates (ADC) are innovative oncology drugs that combine the specificity of monoclonal antibodies and the high efficacy of anticancer payloads, to deliver cytotoxic drugs selectively to cancer cells. Various ADCs were investigated for TNBC and have provided a promise for this aggressive women's cancer including the FDA-approved sacituzumab govitecan. In this chapter, we reviewed different ADCs studied for TNBC including their mechanisms of action, efficacy, and tolerability. Moreover, we have also discussed their therapeutic potential based on combinatorial approaches with other targeted therapies in early and metastatic TNBC.
Collapse
Affiliation(s)
- Ouissam Al Jarroudi
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco.
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco.
| | - Khalid El Bairi
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Said Afqir
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
| |
Collapse
|
21
|
Maecker H, Jonnalagadda V, Bhakta S, Jammalamadaka V, Junutula JR. Exploration of the antibody-drug conjugate clinical landscape. MAbs 2023; 15:2229101. [PMID: 37639687 PMCID: PMC10464553 DOI: 10.1080/19420862.2023.2229101] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 08/31/2023] Open
Abstract
The antibody-drug conjugate (ADC) field has undergone a renaissance, with substantial recent developmental investment and subsequent drug approvals over the past 6 y. In November 2022, ElahereTM became the latest ADC to be approved by the US Food and Drug Administration (FDA). To date, over 260 ADCs have been tested in the clinic against various oncology indications. Here, we review the clinical landscape of ADCs that are currently FDA approved (11), agents currently in clinical trials but not yet approved (164), and candidates discontinued following clinical testing (92). These clinically tested ADCs are further analyzed by their targeting tumor antigen(s), linker, payload choices, and highest clinical stage achieved, highlighting limitations associated with the discontinued drug candidates. Lastly, we discuss biologic engineering modifications preclinically demonstrated to improve the therapeutic index that if incorporated may increase the proportion of molecules that successfully transition to regulatory approval.
Collapse
|
22
|
Chen N, Michaels E, Howard F, Nanda R. The evolving therapeutic landscape of antibody-drug conjugates in breast cancer. Expert Rev Anticancer Ther 2022; 22:1325-1331. [PMID: 36408586 PMCID: PMC9833603 DOI: 10.1080/14737140.2022.2147510] [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] [Received: 09/09/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Antibody-drug conjugates (ADCs) are a relatively new class of anti-cancer therapies approved for a number of malignancies, including breast cancer. Their unique structure, consisting of a monoclonal antibody connected via a linker to a toxic payload, combines characteristics of both targeted therapy and chemotherapy. AREAS COVERED In this review, we discuss the unique molecular structure and pharmacologic principles of ADCs and present the clinical efficacy and relevant toxicities of ADCs both approved and in development. While HER2 is the most studied target with approved agents for both HER2-positive and HER2-low expressing tumors, novel targets in HER2-negative disease have expanded our therapeutic capabilities significantly. EXPERT OPINION ADCs are a promising, novel drug class with significant efficacy in all breast cancer subtypes. They are generally safe and well-tolerated. However, further research is necessary to improve their therapeutic potential. The development of predictive biomarkers to identify patients with greatest benefit, improved understanding of drug resistance to advance combination therapies, and novel targets are needed to further the field.
Collapse
Affiliation(s)
- Nan Chen
- Department of Internal Medicine, University of Chicago
| | | | | | - Rita Nanda
- Department of Internal Medicine, University of Chicago
| |
Collapse
|
23
|
Yazdi SI, Sadeghi M, Saeedzadeh E, Jalilifar M. Radiation dosimetry of 89Zr labeled antibody estimated using the MIRD method and MCNP code. NUCLEAR ENGINEERING AND TECHNOLOGY 2022. [DOI: 10.1016/j.net.2022.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
24
|
Webster BR, Gopal N, Ball MW. Tumorigenesis Mechanisms Found in Hereditary Renal Cell Carcinoma: A Review. Genes (Basel) 2022; 13:2122. [PMID: 36421797 PMCID: PMC9690265 DOI: 10.3390/genes13112122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 09/29/2023] Open
Abstract
Renal cell carcinoma is a heterogenous cancer composed of an increasing number of unique subtypes each with their own cellular and tumor behavior. The study of hereditary renal cell carcinoma, which composes just 5% of all types of tumor cases, has allowed for the elucidation of subtype-specific tumorigenesis mechanisms that can also be applied to their sporadic counterparts. This review will focus on the major forms of hereditary renal cell carcinoma and the genetic alterations contributing to their tumorigenesis, including von Hippel Lindau syndrome, Hereditary Papillary Renal Cell Carcinoma, Succinate Dehydrogenase-Deficient Renal Cell Carcinoma, Hereditary Leiomyomatosis and Renal Cell Carcinoma, BRCA Associated Protein 1 Tumor Predisposition Syndrome, Tuberous Sclerosis, Birt-Hogg-Dubé Syndrome and Translocation RCC. The mechanisms for tumorigenesis described in this review are beginning to be exploited via the utilization of novel targets to treat renal cell carcinoma in a subtype-specific fashion.
Collapse
Affiliation(s)
| | | | - Mark W. Ball
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Antibody-Drug Conjugates in Breast Cancer: What Is Beyond HER2? Cancer J 2022; 28:436-445. [DOI: 10.1097/ppo.0000000000000629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Zhang J, Xia Y, Zhou X, Yu H, Tan Y, Du Y, Zhang Q, Wu Y. Current landscape of personalized clinical treatments for triple-negative breast cancer. Front Pharmacol 2022; 13:977660. [PMID: 36188535 PMCID: PMC9523914 DOI: 10.3389/fphar.2022.977660] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly malignant subtype of breast cancer (BC) with vicious behaviors. TNBC is usually associated with relatively poor clinical outcomes, earlier recurrence, and high propensity for visceral metastases than other BC types. TNBC has been increasingly recognized to constitute a very molecular heterogeneous subtype, which may offer additional therapeutic opportunities due to newly discovered cancer-causing drivers and targets. At present, there are multiple novel targeted therapeutic drugs in preclinical researches, clinical trial designs, and clinical practices, such as platinum drugs, poly ADP-ribose polymerase (PARP) inhibitors, immunocheckpoint inhibitors, androgen receptor inhibitors as well as PI3K/AKT/mTOR targeted inhibitors. These personalized, single, or combinational therapies based on molecular heterogeneity are currently showing positive results. The scope of this review is to highlight the latest knowledge about these potential TNBC therapeutic drugs, which will provide comprehensive insights into the personalized therapeutic strategies and options for combating TNBC.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Yu Xia
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomei Zhou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Honghao Yu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufang Tan
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaying Du
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
De Sanctis R, Jacobs F, Benvenuti C, Gaudio M, Franceschini R, Tancredi R, Pedrazzoli P, Santoro A, Zambelli A. From seaside to bedside: Current evidence and future perspectives in the treatment of breast cancer using marine compounds. Front Pharmacol 2022; 13:909566. [PMID: 36160422 PMCID: PMC9495264 DOI: 10.3389/fphar.2022.909566] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/08/2022] [Indexed: 12/02/2022] Open
Abstract
To date, only few marine natural compounds have been proved to be active in breast cancer (BC). The main marine-derived drugs that have been studied for the treatment of BC are tubulin-binding agents (eribulin and plocabulin), DNA-targeting agents (cytarabine and minor groove binders—trabectedin and lurbinectedin) and Antibody-Drug Conjugates (ADCs). Notably, eribulin is the only approved cytotoxic drug for the treatment of advanced BC (ABC), while cytarabine has a limited indication in case of leptomeningeal diffusion of the disease. Also plocabulin showed limited activity in ABC but further research is needed to define its ultimate potential role. The available clinical data for both trabectedin and lurbinectedin are of particular interest in the treatment of BRCA-mutated tumours and HR deficient disease, probably due to a possible immune-mediated mechanism of action. One of the most innovative therapeutic options for the treatment of BC, particularly in TNBC and HER2-positive BC, are ADCs. Some of the ADCs were developed using a specific marine-derived cytotoxic molecule as payload called auristatin. Among these, clinical data are available on ladiratuzumab vedotin and glembatumumab vedotin in TNBC, and on disitamab vedotin and ALT-P7 in HER2-positive patients. A deeper knowledge of the mechanism of action and of the potential predictive factors for response to marine-derived drugs is important for their rational and effective use, alone or in combination. In this narrative review, we discuss the role of marine-derived drugs for the treatment of BC, although most of them are not approved, and the opportunities that could arise from the potential treasure trove of the sea for novel BC therapeutics.
Collapse
Affiliation(s)
- Rita De Sanctis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Flavia Jacobs
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Chiara Benvenuti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Mariangela Gaudio
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Raul Franceschini
- Department of Chemistry, Università degli studi di Milano Statale, Milan, Italy
| | - Richard Tancredi
- Medical Oncology Unit, ASST Melegnano Martesana, Ospedale A. Uboldo, Milan, Italy
| | - Paolo Pedrazzoli
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- *Correspondence: Alberto Zambelli,
| |
Collapse
|
28
|
Lazaratos AM, Annis MG, Siegel PM. GPNMB: a potent inducer of immunosuppression in cancer. Oncogene 2022; 41:4573-4590. [PMID: 36050467 DOI: 10.1038/s41388-022-02443-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022]
Abstract
The immune system is comprised of both innate and adaptive immune cells, which, in the context of cancer, collectively function to eliminate tumor cells. However, tumors can actively sculpt the immune landscape to favor the establishment of an immunosuppressive microenvironment, which promotes tumor growth and progression to metastatic disease. Glycoprotein-NMB (GPNMB) is a transmembrane glycoprotein that is overexpressed in a variety of cancers. It can promote primary tumor growth and metastasis, and GPNMB expression correlates with poor prognosis and shorter recurrence-free survival in patients. There is growing evidence supporting an immunosuppressive role for GPNMB in the context of malignancy. This review provides a description of the emerging roles of GPNMB as an inducer of immunosuppression, with a particular focus on its role in mediating cancer progression by restraining pro-inflammatory innate and adaptive immune responses.
Collapse
Affiliation(s)
| | - Matthew G Annis
- Goodman Cancer Institute, McGill University, Montréal, QC, Canada.,Department of Medicine, McGill University, Montréal, QC, Canada
| | - Peter M Siegel
- Goodman Cancer Institute, McGill University, Montréal, QC, Canada. .,Department of Medicine, McGill University, Montréal, QC, Canada. .,Department of Biochemistry, McGill University, Montréal, QC, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada. .,Department of Oncology, McGill University, Montréal, QC, Canada.
| |
Collapse
|
29
|
Li Y, Zhang H, Merkher Y, Chen L, Liu N, Leonov S, Chen Y. Recent advances in therapeutic strategies for triple-negative breast cancer. J Hematol Oncol 2022; 15:121. [PMID: 36038913 PMCID: PMC9422136 DOI: 10.1186/s13045-022-01341-0] [Citation(s) in RCA: 353] [Impact Index Per Article: 117.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 01/03/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer (BC) with a poor prognosis. Current treatment options are limited to surgery, adjuvant chemotherapy and radiotherapy; however, a proportion of patients have missed the surgical window at the time of diagnosis. TNBC is a highly heterogeneous cancer with specific mutations and aberrant activation of signaling pathways. Hence, targeted therapies, such as those targeting DNA repair pathways, androgen receptor signaling pathways, and kinases, represent promising treatment options against TNBC. In addition, immunotherapy has also been demonstrated to improve overall survival and response in TNBC. In this review, we summarize recent key advances in therapeutic strategies based on molecular subtypes in TNBC.
Collapse
Affiliation(s)
- Yun Li
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huajun Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yulia Merkher
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia, 141700
| | - Lin Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Na Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia, 141700. .,Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia, 142290.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
30
|
Capici S, Ammoni LC, Meli N, Cogliati V, Pepe FF, Piazza F, Cazzaniga ME. Personalised Therapies for Metastatic Triple-Negative Breast Cancer: When Target Is Not Everything. Cancers (Basel) 2022; 14:cancers14153729. [PMID: 35954393 PMCID: PMC9367432 DOI: 10.3390/cancers14153729] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The purpose of the present review is to shed light on new molecular biomarkers in triple-negative breast cancer (TNBC), showing emerging therapeutic approaches related to specific molecular signatures and their mechanisms of action. A general overview of ongoing clinical trials, future perspectives and differences in approval by American and European regulatory authorities is provided. Abstract Triple-negative breast cancer—defined by the absence of oestrogen/progesterone receptors and human epidermal growth factor receptor 2 expression—is a complex and heterogeneous type of tumour characterised by poor prognosis, aggressive behaviour and lack of effective therapeutic strategies. The identification of new biomarkers and molecular signatures is leading to development of new therapeutic strategies including immunotherapy, targeted therapy and antibody-drug conjugates (ADCs). Against a background where chemotherapy has always been considered the standard of care, evolution towards a precision medicine approach could improve TNBC clinical practice in a complex scenario, with many therapeutic options and new drugs. The aim of this review was to focus on emerging therapeutic targets and their related specific therapy, discussing available and emerging drugs, underlining differences in approval by American and European regulatory authorities and showing the future perspective in the large number of ongoing clinical trials.
Collapse
Affiliation(s)
- Serena Capici
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.); (F.F.P.); (M.E.C.)
| | - Luca Carlofrancesco Ammoni
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.M.); (F.P.)
- Correspondence:
| | - Nicole Meli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.M.); (F.P.)
| | - Viola Cogliati
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.); (F.F.P.); (M.E.C.)
| | - Francesca Fulvia Pepe
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.); (F.F.P.); (M.E.C.)
| | - Francesca Piazza
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.M.); (F.P.)
| | - Marina Elena Cazzaniga
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.); (F.F.P.); (M.E.C.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.M.); (F.P.)
| |
Collapse
|
31
|
Current and New Novel Combination Treatments for Metastatic Triple-Negative Breast Cancer. Curr Oncol 2022; 29:4748-4767. [PMID: 35877237 PMCID: PMC9323790 DOI: 10.3390/curroncol29070377] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has a worse prognosis and remains the most challenging breast cancer subtype to treat. This is largely related to the heterogeneity of this disease and the lack of reliable oncological targets. In this review, we discuss the current standard-of-care treatment options for metastatic TNBC, including recent advances with the use of immunotherapy, PARP inhibitors and antibody-drug conjugates. This review also explores new agents and novel combinations arising in the field for the treatment of advanced TNBC.
Collapse
|
32
|
Dewi C, Fristiohady A, Amalia R, Khairul Ikram NK, Ibrahim S, Muchtaridi M. Signaling Pathways and Natural Compounds in Triple-Negative Breast Cancer Cell Line. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123661. [PMID: 35744786 PMCID: PMC9227697 DOI: 10.3390/molecules27123661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, having a poor prognosis and rapid metastases. TNBC is characterized by the absence of estrogen, progesterone, and human epidermal growth receptor-2 (HER2) expressions and has a five-year survival rate. Compared to other breast cancer subtypes, TNBC patients only respond to conventional chemotherapies, and even then, with limited success. Shortages of chemotherapeutic medication can lead to resistance, pressured index therapy, non-selectivity, and severe adverse effects. Finding targeted treatments for TNBC is difficult owing to the various features of cancer. Hence, identifying the most effective molecular targets in TNBC pathogenesis is essential for predicting response to targeted therapies and preventing TNBC cell metastases. Nowadays, natural compounds have gained attention as TNBC treatments, and have offered new strategies for solving drug resistance. Here, we report a systematic review using the database from Pubmed, Science Direct, MDPI, BioScince, Springer, and Nature for articles screening from 2003 to 2022. This review analyzes relevant signaling pathways and the prospect of utilizing natural compounds as a therapeutic agent to improve TNBC treatments in the future.
Collapse
Affiliation(s)
- Citra Dewi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Pharmacy Department, Faculty of Science and Technology, Mandala Waluya University, Kendari 93561, Indonesia
| | - Adryan Fristiohady
- Faculty of Pharmacy, Halu Oleo University, Kampus Hijau Bumi Tridharma, Kendari 93232, Indonesia;
| | - Riezki Amalia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - Nur Kusaira Khairul Ikram
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Sugeng Ibrahim
- Department of Molecular Biology, Faculty of Medicine, Universitas Katolik Soegijapranata, Semarang 50234, Indonesia;
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Correspondence:
| |
Collapse
|
33
|
Deng S, Leong HC, Datta A, Gopal V, Kumar AP, Yap CT. PI3K/AKT Signaling Tips the Balance of Cytoskeletal Forces for Cancer Progression. Cancers (Basel) 2022; 14:1652. [PMID: 35406424 PMCID: PMC8997157 DOI: 10.3390/cancers14071652] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
The PI3K/AKT signaling pathway plays essential roles in multiple cellular processes, which include cell growth, survival, metabolism, and motility. In response to internal and external stimuli, the PI3K/AKT signaling pathway co-opts other signaling pathways, cellular components, and cytoskeletal proteins to reshape individual cells. The cytoskeletal network comprises three main components, which are namely the microfilaments, microtubules, and intermediate filaments. Collectively, they are essential for many fundamental structures and cellular processes. In cancer, aberrant activation of the PI3K/AKT signaling cascade and alteration of cytoskeletal structures have been observed to be highly prevalent, and eventually contribute to many cancer hallmarks. Due to their critical roles in tumor progression, pharmacological agents targeting PI3K/AKT, along with cytoskeletal components, have been developed for better intervention strategies against cancer. In our review, we first discuss existing evidence in-depth and then build on recent advances to propose new directions for therapeutic intervention.
Collapse
Affiliation(s)
- Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
| | - Hin Chong Leong
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Arpita Datta
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Vennila Gopal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
34
|
Fertal SA, Poterala JE, Ponik SM, Wisinski KB. Stromal Characteristics and Impact on New Therapies for Metastatic Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:1238. [PMID: 35267548 PMCID: PMC8909697 DOI: 10.3390/cancers14051238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/25/2022] Open
Abstract
The heterogenous nature of triple-negative breast cancer (TNBC) is an underlying factor in therapy resistance, metastasis, and overall poor patient outcome. The lack of hormone and growth factor receptors lends to the use of chemotherapy as the first-line treatment for TNBC. However, the failure of chemotherapy demonstrates the need to develop novel immunotherapies, antibody-drug conjugates (ADCs), and other tumor- and stromal-targeted therapeutics for TNBC patients. The potential for stromal-targeted therapy is driven by studies indicating that the interactions between tumor cells and the stromal extracellular matrix (ECM) activate mechanisms of therapy resistance. Here, we will review recent outcomes from clinical trials targeting metastatic TNBC with immunotherapies aimed at programed death ligand-receptor interactions, and ADCs specifically linked to trophoblast cell surface antigen 2 (Trop-2). We will discuss how biophysical and biochemical cues from the ECM regulate the pathophysiology of tumor and stromal cells toward a pro-tumor immune environment, therapy resistance, and poor TNBC patient outcome. Moreover, we will highlight how ECM-mediated resistance is motivating the development of new stromal-targeted therapeutics with potential to improve therapy for this disease.
Collapse
Affiliation(s)
- Shelby A. Fertal
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Johanna E. Poterala
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| | - Suzanne M. Ponik
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Kari B. Wisinski
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| |
Collapse
|
35
|
Actively Targeted Nanomedicines in Breast Cancer: From Pre-Clinal Investigation to Clinic. Cancers (Basel) 2022; 14:cancers14051198. [PMID: 35267507 PMCID: PMC8909490 DOI: 10.3390/cancers14051198] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Despite all the efforts and advances made in the treatment of breast cancer, this pathology continues to be one of the main causes of cancer death in women, particularly triple-negative breast cancer (TNBC), and, although to a lesser degree, HER-2 receptor-positive tumors. Chemotherapy is one of the main treatments available. However, it shows numerous limitations due to its lack of selectivity. In this sense, the selective delivery of antineoplastics to cancer cells can reduce their adverse effects and increase their efficacy. The use of active targeted nanomedicine is a good strategy to achieve this selective chemotherapy. In fact, in recent decades, several active targeted nanoformulations have been approved or reached clinical investigation with excellent results. Among all nanomedicines, antibody-drug conjugates are the most promising. Abstract Breast cancer is one of the most frequently diagnosed tumors and the second leading cause of cancer death in women worldwide. The use of nanosystems specifically targeted to tumor cells (active targeting) can be an excellent therapeutic tool to improve and optimize current chemotherapy for this type of neoplasm, since they make it possible to reduce the toxicity and, in some cases, increase the efficacy of antineoplastic drugs. Currently, there are 14 nanomedicines that have reached the clinic for the treatment of breast cancer, 4 of which are already approved (Kadcyla®, Enhertu®, Trodelvy®, and Abraxane®). Most of these nanomedicines are antibody–drug conjugates. In the case of HER-2-positive breast cancer, these conjugates (Kadcyla®, Enhertu®, Trastuzumab-duocarmycin, RC48, and HT19-MMAF) target HER-2 receptors, and incorporate maytansinoid, deruxtecan, duocarmicyn, or auristatins as antineoplastics. In TNBC these conjugates (Trodelvy®, Glembatumumab-Vedotin, Ladiratuzumab-vedotin, Cofetuzumab-pelidotin, and PF-06647263) are directed against various targets, in particular Trop-2 glycoprotein, NMB glycoprotein, Zinc transporter LIV-1, and Ephrin receptor-4, to achieve this selective accumulation, and include campthotecins, calicheamins, or auristatins as drugs. Apart from the antibody–drug conjugates, there are other active targeted nanosystems that have reached the clinic for the treatment of these tumors such as Abraxane® and Nab-rapamicyn (albumin nanoparticles entrapping placlitaxel and rapamycin respectively) and various liposomes (MM-302, C225-ILS-Dox, and MM-310) loaded with doxorubicin or docetaxel and coated with ligands targeted to Ephrin A2, EPGF, or HER-2 receptors. In this work, all these active targeted nanomedicines are discussed, analyzing their advantages and disadvantages over conventional chemotherapy as well as the challenges involved in their lab to clinical translation. In addition, examples of formulations developed and evaluated at the preclinical level are also discussed.
Collapse
|
36
|
Teicher BA, Morris J. Antibody-Drug Conjugate Targets, Drugs and Linkers. Curr Cancer Drug Targets 2022; 22:463-529. [PMID: 35209819 DOI: 10.2174/1568009622666220224110538] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/22/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
Antibody-drug conjugates offer the possibility of directing powerful cytotoxic agents to a malignant tumor while sparing normal tissue. The challenge is to select an antibody target expressed exclusively or at highly elevated levels on the surface of tumor cells and either not all or at low levels on normal cells. The current review explores 78 targets that have been explored as antibody-drug conjugate targets. Some of these targets have been abandoned, 9 or more are the targets of FDA-approved drugs, and most remain active clinical interest. Antibody-drug conjugates require potent cytotoxic drug payloads, several of these small molecules are discussed, as are the linkers between the protein component and small molecule components of the conjugates. Finally, conclusions regarding the elements for the successful antibody-drug conjugate are discussed.
Collapse
Affiliation(s)
- Beverly A Teicher
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| | - Joel Morris
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| |
Collapse
|
37
|
Biondini M, Kiepas A, El-Houjeiri L, Annis MG, Hsu BE, Fortier AM, Morin G, Martina JA, Sirois I, Aguilar-Mahecha A, Gruosso T, McGuirk S, Rose AAN, Tokat UM, Johnson RM, Sahin O, Bareke E, St-Pierre J, Park M, Basik M, Majewski J, Puertollano R, Pause A, Huang S, Keler T, Siegel PM. HSP90 inhibitors induce GPNMB cell-surface expression by modulating lysosomal positioning and sensitize breast cancer cells to glembatumumab vedotin. Oncogene 2022; 41:1701-1717. [PMID: 35110681 DOI: 10.1038/s41388-022-02206-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/29/2021] [Accepted: 01/20/2022] [Indexed: 12/18/2022]
Abstract
Transmembrane glycoprotein NMB (GPNMB) is a prognostic marker of poor outcome in patients with triple-negative breast cancer (TNBC). Glembatumumab Vedotin, an antibody drug conjugate targeting GPNMB, exhibits variable efficacy against GPNMB-positive metastatic TNBC as a single agent. We show that GPNMB levels increase in response to standard-of-care and experimental therapies for multiple breast cancer subtypes. While these therapeutic stressors induce GPNMB expression through differential engagement of the MiTF family of transcription factors, not all are capable of increasing GPNMB cell-surface localization required for Glembatumumab Vedotin inhibition. Using a FACS-based genetic screen, we discovered that suppression of heat shock protein 90 (HSP90) concomitantly increases GPNMB expression and cell-surface localization. Mechanistically, HSP90 inhibition resulted in lysosomal dispersion towards the cell periphery and fusion with the plasma membrane, which delivers GPNMB to the cell surface. Finally, treatment with HSP90 inhibitors sensitizes breast cancers to Glembatumumab Vedotin in vivo, suggesting that combination of HSP90 inhibitors and Glembatumumab Vedotin may be a viable treatment strategy for patients with metastatic TNBC.
Collapse
Affiliation(s)
- Marco Biondini
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Alex Kiepas
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Physiology, McGill University, Montreal, QC, Canada
| | - Leeanna El-Houjeiri
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Matthew G Annis
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Brian E Hsu
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Anne-Marie Fortier
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Geneviève Morin
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - José A Martina
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Isabelle Sirois
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC, Canada
| | - Adriana Aguilar-Mahecha
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC, Canada
| | - Tina Gruosso
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Shawn McGuirk
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Physiology, McGill University, Montreal, QC, Canada
| | - April A N Rose
- Department of Oncology and Surgery, McGill University, Montreal, QC, Canada
| | - Unal M Tokat
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | | | - Ozgur Sahin
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, USA
| | - Eric Bareke
- Genome Québec Innovation Center, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Morag Park
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Mark Basik
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC, Canada.,Department of Oncology and Surgery, McGill University, Montreal, QC, Canada
| | - Jacek Majewski
- Genome Québec Innovation Center, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Arnim Pause
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Sidong Huang
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | | | - Peter M Siegel
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada. .,Department of Medicine, McGill University, Montreal, QC, Canada. .,Department of Biochemistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
38
|
Abstract
The development of senomorphic drugs to attenuate the senescent phenotype and senolytics to clear pro-inflammatory senescent cells to treat aging-associated disorders is being hotly pursued. The effort is complicated by the fact that senescent cells play a constructive role in some cellular processes such as tissue repair and wound healing. However, concerns about efficacy, which senescent cells to target, and unwanted side effects have created potential roadblocks. Chimeric Antigen Receptor (CAR) T cells directed against urokinase-type plasminogen activator receptor (uPAR), which is expressed on at least a subset of senescent cells (SC) in atherosclerotic plaques and fibrotic livers, removed SC and improved glucose metabolism. A conventional vaccine targeting CD153-expressing senescent T-cells, also improved glucose metabolism in obese mice. Recent work to selectively target senescent cells associated with several pathologies has resulted in the creation of a peptide vaccine that primarily targets endothelial cells expressing high levels of GPNMB, recently identified as a biomarker of senescence. The vaccine reduces atherosclerotic plaque burden and metabolic dysfunction such as glucose intolerance in mouse models of obesity and atherosclerosis. For translation to humans the activity of the vaccine will need to be tightly controlled, as the target, GPNMB has multiple roles in normal physiology including acting to inhibit and possibly resolve inflammation. A promising alternative approach would be to use passive immunization with a monoclonal antibody directed against GPNMB.
Collapse
Affiliation(s)
- Andrew R Mendelsohn
- Panorama Research Institute, 1230 Bordeaux Dr, Sunnyvale, California, United States, 94089.,Regenerative Sciences Institute, 1230 Bordeaux Dr, Sunnyvale, California, United States, 94089;
| | - James Larrick
- Panorama Research Institute, 1230 Bordeaux Drive, Sunnyvale, California, United States, 94089;
| |
Collapse
|
39
|
Salles DC, Asrani K, Woo J, Vidotto T, Liu HB, Vidal I, Matoso A, Netto GJ, Argani P, Lotan TL. GPNMB
expression identifies
TSC1
/2/
mTOR
‐associated and
MiT
family translocation‐driven renal neoplasms. J Pathol 2022; 257:158-171. [PMID: 35072947 PMCID: PMC9310781 DOI: 10.1002/path.5875] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/21/2021] [Accepted: 01/22/2022] [Indexed: 11/20/2022]
Abstract
GPNMB (glycoprotein nonmetastatic B) and other TFE3/TFEB transcriptional targets have been proposed as markers for microphthalmia (MiT) translocation renal cell carcinomas (tRCCs). We recently demonstrated that constitutive mTORC1 activation via TSC1/2 loss leads to increased activity of TFE3/TFEB, suggesting that the pathogenesis and molecular markers for tRCCs and TSC1/2‐associated tumors may be overlapping. We examined GPNMB expression in human kidney and angiomyolipoma (AML) cell lines with TSC2 and/or TFE3/TFEB loss produced using CRISPR–Cas9 genome editing as well as in a mouse model of Tsc2 inactivation‐driven renal tumorigenesis. Using an automated immunohistochemistry (IHC) assay for GPNMB, digital image analysis was employed to quantitatively score expression in clear cell RCC (ccRCC, n = 87), papillary RCC (papRCC, n = 53), chromophobe RCC (chRCC, n = 34), oncocytoma (n = 4), TFE3‐ or TFEB‐driven tRCC (n = 56), eosinophilic solid and cystic RCC (ESC, n = 6), eosinophilic vacuolated tumor (EVT, n = 4), and low‐grade oncocytic tumor (LOT, n = 3), as well as AML (n = 29) and perivascular epithelioid cell tumors (PEComas, n = 8). In cell lines, GPNMB was upregulated following TSC2 loss in a MiT/TFE‐ and mTORC1‐dependent fashion. Renal tumors in Tsc2+/− A/J mice showed upregulation of GPNMB compared with normal kidney. Mean GPNMB expression was significantly higher in tRCC than in ccRCC (p < 0.0001), papRCC (p < 0.0001), and chRCC (p < 0.0001). GPNMB expression in TSC1/2/MTOR alteration‐associated renal tumors (including ESC, LOT, AML, and PEComa) was comparable to that in tRCC. The immunophenotype of tRCC and TSC1/2/MTOR alteration‐associated renal tumors is highly overlapping, likely due to the increased activity of TFE3/TFEB in both, revealing an important caveat regarding the use of TFE3/TFEB‐transcriptional targets as diagnostic markers. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Daniela C. Salles
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Kaushal Asrani
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Juhyung Woo
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Thiago Vidotto
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Hans B. Liu
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Igor Vidal
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Andres Matoso
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - George J. Netto
- Department of Pathology University of Alabama Birmingham Alabama USA
| | - Pedram Argani
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Tamara L. Lotan
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Urology Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Oncology Johns Hopkins University School of Medicine Baltimore MD USA
| |
Collapse
|
40
|
Yang H, Yu X, An Z. Cutaneous Toxicity Associated With Enfortumab Vedotin: A Real-Word Study Leveraging U.S. Food and Drug Administration Adverse Event Reporting System. Front Oncol 2022; 11:801199. [PMID: 35127510 PMCID: PMC8807512 DOI: 10.3389/fonc.2021.801199] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/22/2021] [Indexed: 12/03/2022] Open
Abstract
Introduction Enfortumab vedotin (EV) has been demonstrated to have a significant response rate in early phase trials and is known for its tolerable side-effect profile. Emerging case reports have raised awareness of cutaneous toxicities, which may be a potentially fatal complication. Objective To assess the potential relevance between EV and cutaneous toxicities reports through data mining of the U.S. Food and Drug Administration (FDA) adverse event reporting system (FAERS). Methods Data from January 1, 2019, to November 4, 2021, in the FAERS database were retrieved. Information component (IC) and reporting odds ratio (ROR) were used to evaluate the association between EV and cutaneous toxicities events. Results EV was significantly associated with cutaneous toxicities in the database compared with both all other drugs (ROR 12.90 [10.62–15.66], IC 2.76 [2.52–3.01], middle signal) and platinum-based therapy (ROR 15.11 [12.43–18.37], IC 2.91 [2.66–3.15], middle signal) in the FAERS database. A significant association was detected between EV and all the cutaneous adverse effects (AEs) except erythema, palmar–plantar erythrodysesthesia syndrome, and dermatitis allergic. Both Stevens–Johnson syndrome and toxic epidermal necrolysis occurred 15 times as frequently for EV compared with all other drugs (ROR = 15.20; ROR = 15.52), while Stevens–Johnson syndrome occurred 18 times and toxic epidermal necrolysis occurred 7 times as frequently for EV compared with platinum-based therapy in the database (ROR = 18.74; ROR = 7.80). All groups that limited the gender and age showed a significant association between EV and cutaneous toxicities. Conclusions A significant signal was detected between EV use and cutaneous toxicities. It is worth noting that Stevens–Johnson syndrome and toxic epidermal necrolysis were significantly associated with EV use.
Collapse
|
41
|
Lee S, Cavaliere A, Gallezot JD, Keler T, Michelhaugh SK, Belitzky E, Liu M, Mulnix T, Maher SE, Bothwell ALM, Li F, Phadke M, Mittal S, Marquez-Nostra B. [89Zr]ZrDFO-CR011 positron emission tomography correlates with response to glycoprotein non-metastatic melanoma B-targeted therapy in triple negative breast cancer. Mol Cancer Ther 2022; 21:440-447. [PMID: 35027482 DOI: 10.1158/1535-7163.mct-21-0590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/14/2021] [Accepted: 01/11/2022] [Indexed: 11/16/2022]
Abstract
There is a need for prognostic markers to select patients most likely to benefit from antibody drug conjugate (ADC) therapy. We quantified the relationship between pre-treatment positron emission tomography (PET) imaging of glycoprotein non-metastatic melanoma B (gpNMB) with 89Zr-labeled anti-gpNMB antibody ([89Zr]ZrDFO-CR011) and response to ADC therapy (CDX-011) in triple negative breast cancer (TNBC). First, we compared different PET imaging metrics and found that standardized uptake values (SUV) and tumor-to-heart SUV ratios (SUVR) were sufficient to delineate differences in radiotracer uptake in the tumor of four different cell- and patient-derived tumor models and achieved high standardized effect sizes. These tumor models with varying levels of gpNMB expression were imaged with [89Zr]ZrDFO-CR011 followed by treatment with a single bolus injection of CDX-011. The percent change in tumor volume relative to baseline (% CTV) was then correlated with SUVmean of [89Zr]ZrDFO-CR011 uptake in the tumor. All gpNMB-positive tumor models responded to CDX-011 over 6 weeks of treatment, except one patient-derived tumor re-grew after 4 weeks of treatment. As expected, the gpNMB-negative tumor increased in volume by 130 {plus minus} 59 % at endpoint. The magnitude of pre-treatment SUV had the strongest inverse correlation with the % CTV at 2 - 4 weeks after treatment with CDX-011 (Spearman ρ = -0.8). However, pre-treatment PET imaging with [89Zr]ZrDFO-CR011 did not inform on which tumor types will re-grow over time. Other methods will be needed to predict resistance to treatment.
Collapse
Affiliation(s)
- Supum Lee
- Radiology and Biomedical Imaging, Yale University
| | | | | | | | | | | | - Michael Liu
- Radiology and Biomedical Imaging, Yale University
| | | | | | | | - Fangyong Li
- Yale Center for Analytic Sciences, Yale University School of Public Health
| | - Manali Phadke
- Yale Center for Analytical Sciences, Yale School of Medicine
| | - Sandeep Mittal
- Neurosurgery, Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine
| | | |
Collapse
|
42
|
Ceci C, Lacal PM, Graziani G. Antibody-drug conjugates: Resurgent anticancer agents with multi-targeted therapeutic potential. Pharmacol Ther 2022; 236:108106. [PMID: 34990642 DOI: 10.1016/j.pharmthera.2021.108106] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Antibody-drug conjugates (ADCs) constitute a relatively new group of anticancer agents, whose first appearance took place about two decades ago, but a renewed interest occurred in recent years, following the success of anti-cancer immunotherapy with monoclonal antibodies. Indeed, an ADC combines the selectivity of a monoclonal antibody with the cell killing properties of a chemotherapeutic agent (payload), joined together through an appropriate linker. The antibody moiety targets a specific cell surface antigen expressed by tumor cells and/or cells of the tumor microenvironment and acts as a carrier that delivers the cytotoxic payload within the tumor mass. Despite advantages in terms of selectivity and potency, the development of ADCs is not devoid of challenges, due to: i) low tumor selectivity when the target antigens are not exclusively expressed by cancer cells; ii) premature release of the cytotoxic drug into the bloodstream as a consequence of linker instability; iii) development of tumor resistance mechanisms to the payload. All these factors may result in lack of efficacy and/or in no safety improvement compared to unconjugated cytotoxic agents. Nevertheless, the development of antibodies engineered to remain inert until activated in the tumor (e.g., antibodies activated proteolytically after internalization or by the acidic conditions of the tumor microenvironment) together with the discovery of innovative targets and cytotoxic or immunomodulatory payloads, have allowed the design of next-generation ADCs that are expected to possess improved therapeutic properties. This review provides an overview of approved ADCs, with related advantages and limitations, and of novel targets exploited by ADCs that are presently under clinical investigation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy.
| |
Collapse
|
43
|
Severe cutaneous drug toxicity following enfortumab vedotin treatment for metastatic urothelial carcinoma. JAAD Case Rep 2022; 21:140-143. [PMID: 35242967 PMCID: PMC8857547 DOI: 10.1016/j.jdcr.2022.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
44
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
45
|
Zhai JP, Liu ZH, Wang HD, Huang GL, Man LB. GPNMB overexpression is associated with extensive bone metastasis and poor prognosis in renal cell carcinoma. Oncol Lett 2021; 23:36. [PMID: 34966452 DOI: 10.3892/ol.2021.13154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Glycoprotein non-metastatic protein B (GPNMB) promotes bone metastasis (BM) in various types of cancer. However, GPNMB expression and its function in patients with renal cell carcinoma (RCC) and BM is still unknown. Therefore, the clinical significance of GPNMB and its biological function in RCC with BM was investigated in the present study. A total of 31 patients with RCC and BM were retrospectively collected. The association between GPNMB protein expression level on the primary tumor and the clinicopathological characteristics of the patients was analyzed. Kaplan-Meier analysis was used to investigate the association between GPNMB expression and the prognosis of the patients. The effects of GPNMB inhibition on cell proliferation, migration and invasion in RCC cells were investigated using short hairpin (sh)RNA. High GPNMB expression level was significantly associated with the number (P=0.001) and the extent of BM (P=0.001), Fuhrman grade (P=0.037), and ERK expression level (P=0.003) of the primary tumor. In addition, GPNMB overexpression was significantly associated with poor prognosis with respect to overall survival time (P=0.001). Furthermore, a specific shRNA sequence targeting the GPNMB gene was constructed and transduced into the ACHN cell line, using a lentivirus vector to obtain a stable cell line with low mRNA expression level of GPNMB. Low GPNMB expression level inhibited RCC cell proliferation, which was measured using a Cell Counting Kit-8 assay. Cell migration and invasion ability was significantly decreased in GPNMB knockdown RCC cells compared with that in cells transduced with the negative control shRNA. In addition, the protein expression levels of phosphorylated ERK were lower in the GPNMB shRNA-transduced ACHN cells compared with those in the control cells. Therefore, these results suggested that GPNMB plays an important role in tumor progression in RCC with BM. Furthermore, it might serve as a predictive marker for BM and as a poor prognostic factor in RCC with BM. GPNMB downregulation suppressed the proliferation, migration and invasion of the RCC cells, which may be mediated through the inhibition of the ERK signaling pathway.
Collapse
Affiliation(s)
- Jian-Po Zhai
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| | - Zhen-Hua Liu
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| | - Hai-Dong Wang
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| | - Guang-Lin Huang
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| | - Li-Bo Man
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| |
Collapse
|
46
|
Guo Z, Xing R, Zhao M, Li Y, Lu H, Liu Z. Controllable Engineering and Functionalizing of Nanoparticles for Targeting Specific Proteins towards Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101713. [PMID: 34725943 PMCID: PMC8693047 DOI: 10.1002/advs.202101713] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/15/2021] [Indexed: 05/14/2023]
Abstract
Nanoparticles have been widely used in important biomedical applications such as imaging, drug delivery, and disease therapy, in which targeting toward specific proteins is often essential. However, current targeting strategies mainly rely on surface modification with bioligands, which not only often fail to provide desired properties but also remain challenging. Here an unprecedented approach is reported, called reverse microemulsion-confined epitope-oriented surface imprinting and cladding (ROSIC), for facile, versatile, and controllable engineering coreless and core/shell nanoparticles with tunable monodispersed size as well as specific targeting capability toward proteins and peptides. Via engineering coreless imprinted and cladded silica nanoparticles, the effectiveness and superiority over conventional imprinting of the proposed approach are first verified. The prepared nanoparticles exhibit both high specificity and high affinity. Using quantum dots, superparamagnetic nanoparticles, silver nanoparticles, and upconverting nanoparticles as a representative set of core substrates, a variety of imprinted and cladded single-core/shell nanoparticles are then successfully prepared. Finally, using imprinted and cladded fluorescent nanoparticles as probes, in vitro targeted imaging of triple-negative breast cancer (TNBC) cells and in vivo targeted imaging of TNBC-bearing mice are achieved. This approach opens a new avenue to engineering of nanoparticles for targeting specific proteins, holding great prospects in biomedical applications.
Collapse
Affiliation(s)
- Zhanchen Guo
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Rongrong Xing
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Menghuan Zhao
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Ying Li
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Haifeng Lu
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Zhen Liu
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| |
Collapse
|
47
|
Pestana RC, Roszik J, Groisberg R, Sen S, Van Tine BA, Conley AP, Subbiah V. Discovery of targeted expression data for novel antibody-based and chimeric antigen receptor-based therapeutics in soft tissue sarcomas using RNA-sequencing: clinical implications. Curr Probl Cancer 2021; 45:100794. [PMID: 34656365 DOI: 10.1016/j.currproblcancer.2021.100794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 08/11/2021] [Accepted: 09/02/2021] [Indexed: 11/18/2022]
Abstract
Recent failure of phase 3 trials and paucity of druggable oncogenic drivers hamper developmental therapeutics in sarcomas. Antibody-based therapeutics, like antibody-drug conjugates (ADCs) and chimeric antigen receptor (CAR)-based therapeutics, have emerged as promising strategies for anticancer drug delivery. The efficacy of these novel therapies is highly dependent on expression of the antibody target. We used RNA sequencing data from Cancer Genome Atlas (TCGA) to analyze expression of target antigens in sarcoma subtypes including dedifferentiated liposarcoma (DDLPS; n = 50), uterine leiomyosarcoma (ULMS; n = 27), leiomyosarcoma (STLMS; n = 53), undifferentiated pleomorphic sarcoma (UPS; n = 44), myxofibrosarcoma (MFS; n = 17), synovial sarcoma (SS; n = 10), and malignant peripheral nerve sheath tumor (MPNST; n = 5). We searched published literature and clinicaltrial.gov for ADC targets, bispecific antibodies, immunotoxins, radioimmunoconjugates, SPEAR T-cells, and CAR's that are in clinical trials. CD70 expression was significantly higher in DDLPS, UPS, and MFS than SS and STLMS. CDH3 expression was greater in LMS and ULMS than UPS (P < 0.001), MFS (P < 0.001), and DDLPS (P < 0.001). ERBB2 expression was low; however, it was overexpressed in MPNST when compared with UPS (P < 0.001), and MFS (P < 0.01). GPNMB was highly expressed in most sarcomas, with the exception of SS. LRRC15 also appeared to be a relevant target, especially in UPS. MSLN expression was relatively low except in SS and MPNST. PDGFRA was also highly expressed in most sarcomas with the exception of ULMS and STLMS. TNFRSF8 seems to be most appropriate in DDLPS, as well as MFS. AXL was expressed especially in MFS and STLMS. Sarcoma subtypes express multiple target genes relevant for ADCs, SPEAR T-cells and CAR's, warranting further clinical validation and evaluation.
Collapse
Affiliation(s)
- Roberto Carmagnani Pestana
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas; Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jason Roszik
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roman Groisberg
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas; Rutgers Cancer Institute of New Jersey, New Jersey
| | - Shiraj Sen
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas; Sarah Cannon Research Institute at HealthONE, Denver, Colorado
| | - Brian A Van Tine
- Division of Medical Oncology, Washington University in St. Louis, St Louis, Missouri; Division of Pediatric Hematology and Oncology, St. Louis Children's Hospital, St Louis, Missouri; Siteman Cancer Center, St Louis, Missouri
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
48
|
Khan SA, Sun Z, Dahlberg S, Malhotra J, Keresztes R, Ikpeazu C, Ma P, Ramalingam SS, Pillai R. Efficacy and Safety of Glembatumumab Vedotin in Patients With Advanced or Metastatic Squamous Cell Carcinoma of the Lung (PrECOG 0504). JTO Clin Res Rep 2021; 2:100166. [PMID: 34590018 PMCID: PMC8474292 DOI: 10.1016/j.jtocrr.2021.100166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/07/2021] [Accepted: 03/10/2021] [Indexed: 11/20/2022] Open
Abstract
Introduction Glycoprotein NMB is a transmembrane protein linked with poor prognosis and is expressed in most squamous lung cancer. Glembatumumab vedotin is an antibody-drug conjugate targeting glycoprotein NMB, administered intravenously every 3 weeks in this phase 1 study to determine the safety, tolerability, and maximum tolerated dose in patients who had progressed on any number of previous therapies. Results A total of 13 patients were enrolled; adverse events (of any grade) including dyspnea, neutropenia, respiratory failure, anemia, increased aspartate transaminase/alanine transaminase, diarrhea, and hypophosphatemia were seen in 15% of patients. Grade 5 events included two cases of respiratory failure, either completely or partially attributed to cancer progression. The only other grade 5 event was “disease progression.” The most common adverse events (23%) were decreased appetite, fatigue, rash, and weight loss. The median overall and progression-free survivals were 5.7 months (90% confidence interval: 2.5–16.8) and 2.5 months (90% confidence interval: 1.6–5.8) respectively. Conclusions Glembatumumab vedotin exhibited no serious or unexpected toxicity in this heavily pretreated population, except those caused by disease progression. Modest anticancer activity was observed with a recommendation for a phase 2 dose of 1.9 mg/kg. This portion of the study was not undertaken owing to the company’s decision to discontinue drug development.
Collapse
Affiliation(s)
- Saad A. Khan
- Harold C. Simmons Comprehensive Cancer Center, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Current Affiliation: Cancer Institute, Stanford Medicine, Stanford University, Stanford, California
- Corresponding author. Address for correspondence: Saad A. Khan, MD, Division of Oncology, Department of Medicine, 875 Blake Wilbur Avenue, Stanford, CA 94304.
| | - Zhuoxin Sun
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Suzanne Dahlberg
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, Massachusetts
- Current Affiliation: Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jyoti Malhotra
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Roger Keresztes
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York
| | - Chukwuemeka Ikpeazu
- Division of Medical Oncology, Department of Internal Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Patrick Ma
- Department of Medicine, Penn State University, State College, Pennsylvania
| | - Suresh S. Ramalingam
- Department of Hematology/Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Rathi Pillai
- Department of Hematology/Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
49
|
Reuss JE, Gosa L, Liu SV. Antibody Drug Conjugates in Lung Cancer: State of the Current Therapeutic Landscape and Future Developments. Clin Lung Cancer 2021; 22:483-499. [PMID: 34420859 DOI: 10.1016/j.cllc.2021.07.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022]
Abstract
While both targeted therapy and immunotherapy-based strategies have emerged as frontline standard-of-care for patients with advanced lung cancer, acquired resistance and disease progression remain inevitable in most cases. Chemotherapy is a common salvage option in this scenario, but is limited by a relatively narrow therapeutic index. The emergence of antibody-drug conjugates (ADCs) offer an appealing alternative. ADCs couple the specificity of a monoclonal antibody with the cytotoxic effects of chemotherapy to facilitate the targeted delivery of cytotoxic payloads directly to cancer cells. Here, we review the general structure and function of ADCs, followed by a discussion of emerging ADCs in lung cancer and the future applications of this increasingly relevant class of novel agents.
Collapse
Affiliation(s)
- Joshua E Reuss
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC.
| | - Laura Gosa
- Georgetown University School of Medicine, Georgetown University, Washington, DC
| | - Stephen V Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| |
Collapse
|
50
|
Jakabova A, Bielcikova Z, Pospisilova E, Petruzelka L, Blasiak P, Bobek V, Kolostova K. Characterization of circulating tumor cells in early breast cancer patients receiving neoadjuvant chemotherapy. Ther Adv Med Oncol 2021; 13:17588359211028492. [PMID: 34345252 PMCID: PMC8283058 DOI: 10.1177/17588359211028492] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 06/09/2021] [Indexed: 02/01/2023] Open
Abstract
Background and Aims: The aim of this study was to characterize circulating tumor cells (CTCs) during neoadjuvant chemotherapy (NACT) in early and locally advanced breast cancer (LABC) patients. Using ultrasound, tumor volume measurement was compared with the presence and the molecular nature of CTCs over multiple time intervals corresponding to treatment periods. Methods: A total of 20 patients diagnosed with breast cancer (BC) of different histotypes were monitored during the NACT period and in the follow-up period (~5 years). Peripheral blood for CTCs (n = 115) was taken prior to NACT, after two to three chemotherapy cycles, after the completion of NACT (before surgery) and at some time points during adjuvant therapy. CTCs were enriched using a size-based filtration method (MetaCell®) capturing viable cells, which enabled vital fluorescence microscopy. A set of tumor-associated (TA) genes and chemoresistance-associated (CA) genes was analyzed by qPCR in the enriched CTC fractions. Results: The analysis of tumor volume reduction after administration of anthracyclines (AC) and taxanes (TAX) during NACT showed that AC therapy was responsive in 60% (12/20) of tumors, whereas TAX therapy was responsive in 30% (6/20; n.s.). After NACT, CTCs were still present in 70.5% (12/17) of patients (responders versus non-responders, 61.5% versus 100%; not significant). In triple-negative BC (TNBC) patients (n = 8), tumor volume reduction was observed in 75% cases. CTCs were significantly reduced in 42.9% of all HER2-negative BC patients. In HER2+ tumors, CTC reduction was reported in 16.6% only. Relapses were also more prevalent in the HER2-positive patient group (28.5 versus 66.6%). During NACT, the presence of CTCs (three tests for each patient) identified patients with relapses and indicated significantly shorter progression-free survival (PFS) rates (p = 0.03). Differentiation between progressive disease and non-progressive disease was obtained when the occurrence of excessive expression for CA genes in CTCs was compared (p = 0.024). Absence of tumor volume reduction was also significantly indicative for progressive disease (p = 0.0224). Disseminated CTCs in HER2-negative tumors expressed HER2 in 29% of samples collected during the overall follow-up period (16/55), and in 32% of samples during the follow-up of NACT (10/31). The change accounted for 78.5% of HER2-negative patients (11/14) in total, and 63.6% of the conversion cases occurred during NACT (7/11). For the remaining four patients (36.3%), conversion to HER2+ CTCs occurred later during adjuvant therapy. We believe there is the possibility of preventing further progression by identifying less responsive tumors during NACT using CTC monitoring, which could also be used effectively during adjuvant therapy.
Collapse
Affiliation(s)
- Anna Jakabova
- Radiotherapy and Oncology Clinic, Laboratory of Personalized Medicine, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Zuzana Bielcikova
- Department of Oncology, First Faculty of Medicine Charles University and General University Hospital in Prague, Czech Republic
| | - Eliska Pospisilova
- Radiotherapy and Oncology Clinic, Laboratory of Personalized Medicine, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, First Faculty of Medicine Charles University and General University Hospital in Prague, Czech Republic
| | - Piotr Blasiak
- Department and Clinic of Thoracic Surgery, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Vladimir Bobek
- Radiotherapy and Oncology Clinic, Laboratory of Personalized Medicine, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Katarina Kolostova
- Radiotherapy and Oncology Clinic, Laboratory of Personalized Medicine, University Hospital, Kralovske Vinohrady, Srobarova 50, Prague, 100 34, Czech Republic
| |
Collapse
|