1
|
Rossi V, Turati A, Rosato A, Carpanese D. Sacituzumab govitecan in triple-negative breast cancer: from bench to bedside, and back. Front Immunol 2024; 15:1447280. [PMID: 39211043 PMCID: PMC11357913 DOI: 10.3389/fimmu.2024.1447280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Triple-negative breast cancer (TNBC) represents a major therapeutic challenge due to its heterogeneous and aggressive phenotype, and limited target-specific treatment options. The trophoblast cell surface antigen (Trop-2), a transmembrane glycoprotein overexpressed in various cancers, has emerged as a promising target for TNBC. Sacituzumab govitecan (SG), an antibody-drug conjugate (ADC) that targets Trop-2, has recently entered treatment algorithms for advanced and metastatic TNBC, independently from Trop-2 expression status, with manageable toxicity. Despite the impressive results, questions remain unsolved regarding its efficacy, safety profile, and Trop-2 biological role in cancer. Currently, Trop-2 cannot be designated as a predictive biomarker in SG treatment, albeit its expression correlates with disease outcome, yet its levels are not uniform across all TNBCs. Additionally, data regarding Trop-2 expression variations in primary and metastatic sites, and its interplay with other biomarkers are still ambiguous but mandatory in light of future applications of SG in other indications and settings. This poses the questions of a careful evaluation of the efficacy and toxicity profile of SG in such early stages of disease, and in personalized and combinatorial strategies. Research and clinical data are mandatory to address SG drawbacks and minimize its benefits, to realize its full potential as therapeutic agent in different epithelial tumors.
Collapse
Affiliation(s)
- Valentina Rossi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| | - Alessandra Turati
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Debora Carpanese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| |
Collapse
|
2
|
Chiba Y, Kojima Y, Yazaki S, Yoshida H, Takamizawa S, Kitadai R, Saito A, Okuma HS, Nishikawa T, Shimoi T, Sudo K, Noguchi E, Uno M, Ishikawa M, Kato T, Fujiwara Y, Yonemori K. Trop-2 expression and the tumor immune microenvironment in cervical cancer. Gynecol Oncol 2024; 187:51-57. [PMID: 38723340 DOI: 10.1016/j.ygyno.2024.04.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/02/2024] [Accepted: 04/23/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Trophoblast Cell Surface Antigen 2 (Trop-2) is a transmembrane glycoprotein that is overexpressed in various cancers, with immunological significance as a target for tumor-reactive T-cells. We aimed to investigate the association between the expression of Trop-2 and the tumor immune microenvironment in cervical cancer. METHODS The study included 123 patients with cervical cancer who underwent primary surgery between 2000 and 2020 in our hospital. Trop-2 expression was evaluated using anti-Trop-2 monoclonal antibody clone MAB650. Immune biomarkers, including PD-L1 (22C3), CD3 (PS1), and CD8 (4B11), were also evaluated. Trop-2 and PD-L1 positivity were defined by an H-score ≥ 10 and a combined positive score (CPS) ≥1, respectively. Tumor-infiltrating lymphocytes (TILs) were assessed in the five selected independent areas. The correlation between Trop-2 expression and immune biomarkers was analyzed. RESULTS The cohort comprised patients with squamous cell carcinoma (SCC) (54.5%) and non-SCC (45.5%). Trop-2 was positive in 84.6% of samples and more commonly expressed in SCC (SCC vs. non-SCC; 97.0% vs. 69.6%, p < 0.001). Intratumoral CD3+ and CD8 + TILs were significantly more common in Trop-2-positive cases (CD3, Mann-Whitney U = 383, p < 0.0001; CD8, U = 442, p < 0.0001). Additionally, significant positive correlations were found between the Trop-2 H-score and immune markers (CD3 + TILs, r = 0.295, p < 0.001; CD8 + TILs, r = 0.267, p = 0.001; PD-L1 CPS, r = 0.178, p = 0.025). No significant associations were detected between TILs and other clinicopathological features, including prognosis. CONCLUSION Expression of Trop-2 in cervical cancer is associated with increased levels of intratumoral TILs, indicating the potential of Trop-2 targeted therapy alone or in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Yohei Chiba
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Yuki Kojima
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Shu Yazaki
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Hiroshi Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan.
| | | | - Rui Kitadai
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Ayumi Saito
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | | | - Tadaaki Nishikawa
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Kazuki Sudo
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Emi Noguchi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Masaya Uno
- Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan.
| | - Mitsuya Ishikawa
- Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan.
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan.
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| |
Collapse
|
3
|
Zhai C, Cui Y, Guo L, Chen C, Song Y, Zhong J, Wang Y. Progress in the study of antibody-drug conjugates for the treatment of cervical cancer. Front Oncol 2024; 14:1395784. [PMID: 38903711 PMCID: PMC11187480 DOI: 10.3389/fonc.2024.1395784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
Cervical cancer is the second most prevalent malignancy affecting women's health globally, and the number of morbidity and mortality from cervical cancer continues to rise worldwide. The 5-year survival rate of patients with recurrent or metastatic cervical cancer is significantly reduced, and existing treatment modalities have low efficacy and high adverse effects, so there is a strong need for new, effective, and well-tolerated therapies. Antibody-drug conjugates (ADCs) are a new targeted therapeutic modality that can efficiently kill tumor cells. This review aims to summarize the composition, research, and development history and mechanism of action of ADCs, to review the research progress of ADCs in the treatment of cervical cancer, and to summarize and prospect the application of ADCs.
Collapse
Affiliation(s)
- Congcong Zhai
- Department of Oncology, Gannan Medical University, Ganzhou, China
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| | - Yan Cui
- Department of Oncology, Bengbu Medical University, Lu’an, China
| | - Ling Guo
- Department of Oncology, Gannan Medical University, Ganzhou, China
| | - Cixiang Chen
- Department of Oncology, Gannan Medical University, Ganzhou, China
| | - Yanfang Song
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| | - Jinghua Zhong
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Yili Wang
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| |
Collapse
|
4
|
Greenman M, McNamara B, Mutlu L, Santin AD. Entering the golden age for antibody-drug conjugates in gynecologic cancer. Oncoscience 2024; 11:51-52. [PMID: 38770444 PMCID: PMC11104406 DOI: 10.18632/oncoscience.604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Indexed: 05/22/2024] Open
Affiliation(s)
- Michelle Greenman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Levent Mutlu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Alessandro D. Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
5
|
Mallmann MR, Tamir S, Alfter K, Ratiu D, Quaas A, Domroese CM. Expression of Potential Antibody-Drug Conjugate Targets in Cervical Cancer. Cancers (Basel) 2024; 16:1787. [PMID: 38730739 PMCID: PMC11083445 DOI: 10.3390/cancers16091787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024] Open
Abstract
(1) Background: There is a huge unmet clinical need for novel treatment strategies in advanced and recurrent cervical cancer. Several cell membrane-bound molecules are up-regulated in cancer cells as compared to normal tissue and have revived interest with the introduction of antibody-drug conjugates (ADCs). (2) Methods: In this study, we characterize the expression of 10 potential ADC targets, TROP2, mesotheline, CEACAM5, DLL3, folate receptor alpha, guanylatcyclase, glycoprotein NMB, CD56, CD70 and CD138, on the gene expression level. Of these, the three ADC targets TROP2, CEACAM5 and CD138 were further analyzed on the protein level. (3) Results: TROP2 shows expression in 98.5% (66/67) of cervical cancer samples. CEACAM5 shows a stable gene expression profile and overall, 68.7% (46/67) of cervical cancer samples are CEACAM-positive with 34.3% (23/67) of cervical cancer samples showing at least moderate or high expression. Overall, 73.1% (49/67) of cervical cancer samples are CD138-positive with 38.8% (26/67) of cervical cancer samples showing at least moderate or high expression. (4) Conclusions: TROP2, CEACAM5 or CD138 do seem suitable for further clinical research and the data presented here might be used to guide further clinical trials with ADCs in advanced and recurrent cervical cancer patients.
Collapse
Affiliation(s)
- Michael R. Mallmann
- Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; (D.R.); (A.Q.); (C.M.D.)
- Department of Obstetrics and Gynecology, University of Cologne, 50931 Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Germany
- Department of Obstetrics and Gynecology, Hospital of the City of Cologne, 51069 Cologne, Germany; (S.T.); (K.A.)
| | - Sina Tamir
- Department of Obstetrics and Gynecology, Hospital of the City of Cologne, 51069 Cologne, Germany; (S.T.); (K.A.)
| | - Katharina Alfter
- Department of Obstetrics and Gynecology, Hospital of the City of Cologne, 51069 Cologne, Germany; (S.T.); (K.A.)
| | - Dominik Ratiu
- Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; (D.R.); (A.Q.); (C.M.D.)
- Department of Obstetrics and Gynecology, University of Cologne, 50931 Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Germany
| | - Alexander Quaas
- Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; (D.R.); (A.Q.); (C.M.D.)
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Germany
- Department of Pathology, University of Cologne, 50931 Cologne, Germany
| | - Christian M. Domroese
- Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; (D.R.); (A.Q.); (C.M.D.)
- Department of Obstetrics and Gynecology, University of Cologne, 50931 Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Germany
| |
Collapse
|
6
|
Sathe AG, Singh I, Singh P, Diderichsen PM, Wang X, Chang P, Taqui A, Phan S, Girish S, Othman AA. Population Pharmacokinetics of Sacituzumab Govitecan in Patients with Metastatic Triple-Negative Breast Cancer and Other Solid Tumors. Clin Pharmacokinet 2024; 63:669-681. [PMID: 38578394 PMCID: PMC11106201 DOI: 10.1007/s40262-024-01366-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND AND OBJECTIVE Sacituzumab govitecan (SG) is an antibody-drug conjugate composed of an antibody with affinity for Trop-2 coupled to SN-38 via hydrolyzable linker. SG is approved for patients with metastatic triple-negative breast cancer (mTNBC) who have received two or more prior chemotherapies (at least one in a metastatic setting) and for patients with pretreated hormone receptor positive (HR+)/human epidermal growth factor receptor 2 negative (HER2-) metastatic breast cancer. METHODS In these analyses, the pharmacokinetics of SG, free SN-38, and total antibody (tAB) were characterized using data from 529 patients with mTNBC or other solid tumors across two large clinical trials (NCT01631552; ASCENT, NCT02574455). Three population pharmacokinetic models were constructed using non-linear mixed-effects modeling; clinically relevant covariates were evaluated to assess their impact on exposure. Models for SG and tAB were developed independently whereas free SN-38 was sequentially generated via a first-order release process from SG. RESULTS Pharmacokinetics of the three analytes were each described by a two-compartment model with estimated body weight-based scaling exponents for clearance and volume. Typical parameter estimates for clearance and steady-state volume of distribution were 0.133 L/h and 3.68 L for SG and 0.0164 L/h and 4.26 L for tAB, respectively. Mild-to-moderate renal impairment, mild hepatic impairment, age, sex, baseline albumin level, tumor type, UGT1A1 genotype, or Trop-2 expression did not have a clinically relevant impact on exposure for any of the three analytes. CONCLUSIONS These analyses support the approved SG dosing regimen of 10 mg/kg as intravenous infusion on days 1 and 8 of 21-day cycles and did not identify a need for dose adjustment based on evaluated covariates or disease characteristics.
Collapse
Affiliation(s)
- Abhishek G Sathe
- Clinical Pharmacology, Gilead Sciences, Inc., 333 Lakeside Dr., Foster City, CA, 94404, USA
| | - Indrajeet Singh
- Clinical Pharmacology, Gilead Sciences, Inc., 333 Lakeside Dr., Foster City, CA, 94404, USA
| | - Pratap Singh
- Clinical Pharmacology, Gilead Sciences, Inc., 333 Lakeside Dr., Foster City, CA, 94404, USA
| | - Paul M Diderichsen
- Integrated Drug Development Consulting, Certara USA, Inc., Princeton, NJ, USA
| | - Xiaohui Wang
- Integrated Drug Development Consulting, Certara USA, Inc., Princeton, NJ, USA
| | - Peter Chang
- Integrated Drug Development Consulting, Certara USA, Inc., Princeton, NJ, USA
| | - Atiya Taqui
- Clinical Pharmacology, Gilead Sciences, Inc., 333 Lakeside Dr., Foster City, CA, 94404, USA
| | - See Phan
- Clinical Research, Gilead Sciences, Inc., Foster City, CA, USA
| | - Sandhya Girish
- Clinical Pharmacology, Gilead Sciences, Inc., 333 Lakeside Dr., Foster City, CA, 94404, USA
| | - Ahmed A Othman
- Clinical Pharmacology, Gilead Sciences, Inc., 333 Lakeside Dr., Foster City, CA, 94404, USA.
| |
Collapse
|
7
|
Mertens RB, Makhoul EP, Li X, Dadmanesh F. Comparative expression of trophoblast cell-surface antigen 2 (TROP2) in the different molecular subtypes of invasive breast carcinoma: An immunohistochemical study of 94 therapy-naive primary breast tumors. Ann Diagn Pathol 2024; 68:152226. [PMID: 37995412 DOI: 10.1016/j.anndiagpath.2023.152226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Sacituzumab govitecan, targeting trophoblast cell-surface antigen 2 (TROP2), is approved for the treatment of triple-negative and hormone receptor-positive/HER2-negative breast cancers. However, detailed studies comparing TROP2 protein expression in the different molecular subtypes of breast cancer are limited, and definitive evidence supporting the use of TROP2 as a biomarker for predicting response to this agent in patients with breast cancer is currently lacking. OBJECTIVE To compare the expression of TROP2 in the different molecular subtypes of breast cancer. METHODS Immunohistochemical staining for TROP2 was performed on 94 therapy-naive primary invasive breast carcinomas, including 25 luminal A-like, 25 luminal B-like, 19 HER2-like, and 25 triple-negative tumors. RESULTS Intermediate to high levels of TROP2 expression were observed in the majority of carcinomas of each molecular subtype, with a wide range of expression in each subtype. Occasional tumors with low or absent TROP2 expression were encountered, including two metaplastic carcinomas which were completely negative for TROP2. CONCLUSIONS Our observations support the continued investigation of the efficacy of sacituzumab govitecan in all molecular subtypes of breast carcinoma. Furthermore, the observed wide range of expression of TROP2 suggests that TROP2 may have potential utility as a biomarker for predicting responsiveness to sacituzumab govitecan. If this proves to be the case, then immunohistochemical staining for TROP2 would be critical for identifying those patients whose tumors are completely negative for TROP2, since these patients may be least likely or unlikely to respond to this agent, and alternative therapies may be more appropriate in such instances.
Collapse
Affiliation(s)
- Richard B Mertens
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA.
| | - Elias P Makhoul
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Xiaomo Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Farnaz Dadmanesh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| |
Collapse
|
8
|
Poorebrahim M, Quiros-Fernandez I, Marmé F, Burdach SE, Cid-Arregui A. A costimulatory chimeric antigen receptor targeting TROP2 enhances the cytotoxicity of NK cells expressing a T cell receptor reactive to human papillomavirus type 16 E7. Cancer Lett 2023; 566:216242. [PMID: 37217069 DOI: 10.1016/j.canlet.2023.216242] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/11/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023]
Abstract
Immune cells modified to express a tumor-reactive T cell receptor (TCR) have shown limited efficacy as stand-alone therapy against solid tumors. Genital and oropharyngeal carcinomas induced by human papillomavirus (HPV) type 16 express constitutively its E6 and E7 oncoproteins, which makes them convenient targets for adoptive cell immunotherapy. However, viral antigen presentation by tumor cells is low and limits the anti-tumor efficacy of CD8+ T cells. To enhance the functionality of immune effector cells, we have devised a strategy combining a costimulatory chimeric antigen receptor (CAR) with a TCR. We used a clinically tested TCR specific to E7 (E7-TCR) of HPV16 and a newly constructed CAR targeting the trophoblast cell surface antigen 2 (TROP2), which carried the intracellular costimulatory domains CD28 and 4-1BB, but was devoid of the CD3ζ domain. Flow cytometry analyses showed a notable upregulation of activation markers and of cytolytic molecule release by NK-92 cells genetically engineered to express CD3, CD8 and both E7-TCR and TROP2-CAR, after co-incubation with HPV16+ cervical cancer cells. Furthermore, the E7-TCR/TROP2-CAR NK-92 cells demonstrated enhanced antigen-specific activation and augmented cytotoxicity against tumor cells compared with NK-92 cells expressing the E7-TCR alone. A costimulatory TROP2-CAR can synergistically cooperate with the E7-TCR in NK cells thereby enhancing their signaling strength and antigen-specific cytotoxicity. This approach might improve the outcome of adoptive cell immunotherapies for HPV16+ cancer patients that are currently under investigation.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany; Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, Canada
| | - Isaac Quiros-Fernandez
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany; Research Center on Tropical Diseases (CIET)/Research Center on Surgery and Cancer (CICICA), Faculty of Microbiology, Universidad de Costa Rica, San Jose, Costa Rica
| | - Frederik Marmé
- Department of Obstetrics and Gynecology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Eg Burdach
- Translational Pediatric Cancer Research Action - Institute of Pathology, TUM School of Medicine - Technical University of Munich, Trogerstr. 22 - RG - 540.0.11, D-81675, München, Germany; Department of Molecular Oncology, British Columbia Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Angel Cid-Arregui
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
9
|
Condic M, Egger EK, Klümper N, Kristiansen G, Mustea A, Thiesler T, Ralser DJ. TROP-2 is widely expressed in vulvar squamous cell carcinoma and represents a potential new therapeutic target. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04761-8. [PMID: 37067548 PMCID: PMC10374825 DOI: 10.1007/s00432-023-04761-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/08/2023] [Indexed: 04/18/2023]
Abstract
PURPOSE Vulvar squamous cell carcinoma (VSCC) is a rare malignancy of the female genital tract with increasing incidence rates. Etiologically, HPV-dependent and HPV-independent VSCC are distinguished. Surgical treatment and/or radiotherapy represent the therapeutic mainstay for localized disease. For recurrent or metastatic VSCC, treatment options are limited. Research has identified trophoblast cell surface antigen 2 (TROP-2) to be broadly expressed across different tumor entities. The aim of the present study was to systematically investigate the expression of TROP-2 in VSCC. METHODS TROP-2 protein expression was investigated by immunohistochemistry in a cohort comprising n = 103 patients with primary VSCC. A four-tier scoring system (0: no staining, 1 + : low staining, 2 + : moderate staining, 3 + : high staining) was applied for quantification of protein expression. For further analyses, two groups (low TROP-2 expression: 0/1 + ; high TROP-2 expression: 2 + /3 +) were generated. The entire study cohort, as well as HPV-dependent and HPV-independent VSCC were considered separately. RESULTS In the entire VSCC study cohort, TROP-2 expression was present in 97.1% of all cases (n = 100) with 74.8% displaying high TROP-2 expression (2 + /3 +). Only 2.9% of tumors showed absent TROP-2 expression. Of note, all HPV-dependent VSCC (n = 18) demonstrated high TROP-2 expression (2 + /3 +). In the subgroup of HPV-independent VSCC (n = 70), high TROP-2 expression was associated with favorable clinical outcomes based on log rank test and univariate cox analysis. CONCLUSION TROP-2 protein expression is of prognostic value in HPV-independent VSCC. The broad expression of TROP-2 in VSCC indicates the TROP-2 directed ADC Sacituzumab govitecan as a potential new therapeutic strategy for VSCC patients.
Collapse
Affiliation(s)
- Mateja Condic
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany
| | - Eva K Egger
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany
| | - Niklas Klümper
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
| | | | - Alexander Mustea
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany
| | - Thore Thiesler
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Damian J Ralser
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
10
|
Xing Y, Yasinjan F, Du Y, Geng H, Zhang Y, He M, Guo R, Yang L, Cui J, Mu D, Liu Z, Wang H. Immunotherapy in cervical cancer: From the view of scientometric analysis and clinical trials. Front Immunol 2023; 14:1094437. [PMID: 36817443 PMCID: PMC9935705 DOI: 10.3389/fimmu.2023.1094437] [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: 11/10/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Background Cervical cancer is the fourth most cancer and the fourth leading cause of cancer-related deaths in women worldwide. Current treatment for patients with advanced cervical cancer is limited. And in the urgent demand for novel effective therapies both as the first and the second line treatment for these patients, immunotherapy is developing fast and has made some achievements. Methods This study incorporated 1,255 topic-related articles and reviews from 1999 to 2022 in the Web of Science Core Collection (WoSCC). The WoS platform, Citespace, and VOS viewer provided the annual distribution of publications and citations, the analysis of researching countries and institutions, references, keywords (co-occurrence analysis, burst analysis, and timeline view analysis), and researching authors, respectively. For clinical trials, 720 trials and 114 trials from ClinicalTrials.gov and ICTRP were retrieved, respectively. And 296 trials were finally incorporated into the analysis. Results The scientometric analysis showed that the study of immunotherapies in cervical cancer developed fast in recent years. Most publications were from the United States, followed by China. Seven of the top 10 co-cited references belong to clinical trials, and five of them were published in recent five years. There are lots of clinical trials us specific treatment patterns, some of which have represented excellent effects. Conclusions Both the scientometric analysis of the 1,255 publications and the analysis of clinical trials showed that the field of immunotherapies in cervical cancer developed so fast in recent years. It was found that a lot of clinical trials using various immunotherapies (mainly vaccine therapy, adoptive cell therapy, immune checkpoint blockade, and antibody-drug conjugate) for advanced cervical cancer are currently ongoing or have represented considerable effect. Centered in immunotherapies, immune checkpoint blockades have represented great efficacy and huge potential, especially combined with other therapies such as chemotherapy, targeted therapy, and other immunotherapies.
Collapse
Affiliation(s)
- Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Feroza Yasinjan
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yajie Du
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Huayue Geng
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Minghua He
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jiayue Cui
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Dongmei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Ziling Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Tymon-Rosario J, Gorman M, Richardson DL, Washington C, Santin AD. Advances in antibody-drug conjugates for gynecologic malignancies. Curr Opin Obstet Gynecol 2023; 35:6-14. [PMID: 36484278 DOI: 10.1097/gco.0000000000000838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) represent a new class of drugs that combine a surface receptor-targeting antibody linked to a cytotoxic molecule delivering the potent cytotoxic payload directly to tumor cells. This review summarizes the current literature demonstrating their use in the treatment of gynecologic malignancies. RECENT FINDINGS Tisotumab vedotin is the first U.S. Food and Drug Administration (FDA) approved ADC for the treatment of gynecologic cancers. While in the phase 3 randomized controlled trial in platinum resistant ovarian cancer patients, FORWARD 1, mirvetuximab did not meet its primary endpoint of progression-free survival. But we await more recent data from the two ongoing phase 3 trials of mirvetuximab in recurrent ovarian cancer patients. HER2/neu, Napi2b, mesothelin, and human trophoblast cell-surface marker (Trop-2) overexpression have also been exploited as excellent targets by novel ADCs in multiple tumors including ovarian, endometrial, and cervical cancers. SUMMARY Current evidence strongly supports the use of ADCs and ongoing clinical trials will provide further information into the potential of making these drugs part of current standard practice allowing patients to be treated with a higher level of personalized cancer care.
Collapse
Affiliation(s)
- Joan Tymon-Rosario
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New Hyde Park, New York 11040
| | - Megan Gorman
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New Hyde Park, New York 11040
| | - Debra L Richardson
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, Connecticut 06520
| | - Christina Washington
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, Connecticut 06520
| | - Alessandro D Santin
- Stephenson Cancer Center at the University of Oklahoma Health Sciences Center, 800 N.E. 10 Street, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
12
|
Liu X, Deng J, Yuan Y, Chen W, Sun W, Wang Y, Huang H, Liang B, Ming T, Wen J, Huang B, Xing D. Advances in Trop2-targeted therapy: Novel agents and opportunities beyond breast cancer. Pharmacol Ther 2022; 239:108296. [PMID: 36208791 DOI: 10.1016/j.pharmthera.2022.108296] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022]
Abstract
Trop2 is a transmembrane glycoprotein and calcium signal transducer with limited expression in normal human tissues. It is consistently overexpressed in a variety of malignant tumors and participates in several oncogenic signaling pathways that lead to tumor development, invasion, and metastasis. As a result, Trop2 has become an attractive therapeutic target in cancer treatment. The anti-Trop2 antibody-drug conjugate (Trodelvy™, sacituzumab govitecan) has been approved to treat metastatic triple-negative breast cancer. However, it is still unclear whether the success observed in Trop2-positive breast cancer could be replicated in other tumor types, owing to the differences in the expression levels and functions of Trop2 across cancer types. In this review, we summarize the recent progress on the structures and functions of Trop2 and highlight the potential diagnostic and therapeutic value of Trop2 beyond breast cancer. In addition, the promising novel Trop2-targeted agents in the clinic were discussed, which will likely alter the therapeutic landscape of Trop2-positive tumors in the future.
Collapse
Affiliation(s)
- Xinlin Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Junwen Deng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Yang Yuan
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Wenshe Sun
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Yanhong Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Haiming Huang
- Shanghai Asia United Antibody Medical Co., Ltd, Shanghai 201203, China
| | - Bing Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Tao Ming
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100094, China
| | - Jialian Wen
- School of Social Science, The University of Manchester, Manchester, UK
| | - Binghuan Huang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
13
|
Abstract
OBJECTIVE To evaluate the antibody-drug conjugate- sacituzumab govitecan, its pharmacological properties, toxicity, data supporting efficacy against a wide variety of solid tumors beyond breast cancer, and potential future uses. DATA SOURCES Articles in MEDLINE/PubMed databases and the National Institutes of Health Clinical Trials Registry (http://www. clinicaltrials.gov) between January 1, 2015, and July 1, 2021 using MeSH terms sacituzumab govitecan(- hziy) and solid tumors were reviewed. DATA SUMMARY Antibody-drug conjugates (ADC's) are a subclass of emerging cancer therapeutics which combines chemotherapy with targeted antibodies. Sacituzumab govitecan (SG) is a novel antibody drug conjugate that has recently been approved by the Food and Drug Administration (FDA) in adult patients for the treatment of unresectable locally advanced or metastatic triple-negative breast cancer (mTNBC) who have received two or more prior systemic therapies, at least one of them for metastatic disease. The approval of sacituzumab govitecan provides a new option for solid tumors that need to be further explored. In this review article, we discussed the pharmacokinetics, pharmacodynamics, safety profile of sacituzumab govitecan and various ongoing clinical trials on sacituzumab govitecan. CONCLUSION Sacituzumab is a significant advancement made in cancer therapy. SG has showed significantly improved Health-related quality of life (HRQoL) in addition to prolonged progression free survival and Over all survival in addition to maintaining a good safety profile. Multiple clinical trials on SG are ongoing to evaluate the potential use of SG as neoadjuvant therapy in triple negative breast cancer, as an Adjuvant therapy, in combination with immunotherapy, and also for various solid tumors.
Collapse
Affiliation(s)
- Sindhusha Veeraballi
- Department of Medical Education, 22423Saint Michael's Medical Center, New York Medical College, New Jersey, United States
| | - Zaineb Khawar
- Medical student, 136414Saint Gorge's University school of medicine, St George's, Grenada
| | - Hafiz Muhammad Aslam
- Department of Hematology/Oncology, 3627East carolina university, Greenville, North Carolina, United States
| | - Mahvish Muzaffar
- Department of Hematology/Oncology, 3627East carolina university, Greenville, North Carolina, United States
| |
Collapse
|
14
|
The European Medicines Agency review of sacituzumab govitecan for the treatment of triple-negative breast cancer. ESMO Open 2022; 7:100497. [PMID: 35642987 PMCID: PMC9149193 DOI: 10.1016/j.esmoop.2022.100497] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 11/24/2022] Open
Abstract
Sacituzumab govitecan (SG) is an antineoplastic agent which combines a humanized monoclonal antibody binding to trophoblast cell surface antigen-2 (Trop-2)-expressing cancer cells, linked with cytotoxic moiety SN-38 (govitecan) with topoisomerase I inhibitor action. On 22 November 2021, a marketing authorization valid through the European Union (EU) was issued under the European Medicines Agency (EMA)’s accelerated assessment program for SG as monotherapy for the treatment of adult patients with unresectable or metastatic triple-negative breast cancer (mTNBC) who have received two or more prior systemic therapies, including at least one of them for advanced disease. The assessment was based on results from an open-label, randomized, phase III trial to evaluate the safety, tolerability, pharmacokinetics and efficacy of SG versus treatment of physician’s choice (TPC) in patients with mTNBC who received at least two prior treatments including at least one of them for advanced disease. The efficacy results in the overall population, based on mature data, showed a statistically significant improvement of SG over TPC in progression-free survival (PFS) and overall survival (OS). The median PFS was 4.8 months versus 1.7 months [hazard ratio (HR) = 0.43, n = 529; 95% CI 0.35-0.54; P < 0.0001] and the median OS was 11.8 months versus 6.9 months (HR = 0.51, n = 529; 95% CI 0.41-0.62; P < 0.0001). The most common (>30%) side effects of SG were diarrhea, neutropenia, nausea, fatigue, alopecia, anemia, constipation and vomiting. The aim of this manuscript is to summarize the scientific review of the application leading to regulatory approval in the EU. Trodelvy (SG) received a marketing authorization valid throughout the EU on 22 November 2021. SG is indicated for adults with unresectable or metastatic TBNC. SG is an antibody–drug conjugate considered a first-in-class medicine. SG prolonged OS and PFS ∼5 and 3 months, respectively, when compared to TPC. The most common serious side effects are febrile neutropenia and diarrhea.
Collapse
|
15
|
Trop-2 in Upper Tract Urothelial Carcinoma. Curr Oncol 2022; 29:3911-3921. [PMID: 35735421 PMCID: PMC9222112 DOI: 10.3390/curroncol29060312] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 11/19/2022] Open
Abstract
Trophoblast cell surface antigen 2 (Trop-2, encoded by TACSTD2) is the target protein of sacituzumab govitecan, a novel antibody-drug conjugate for locally advanced or metastatic urothelial carcinoma. However, the expression status of Trop-2 in upper tract urothelial carcinoma (UTUC) remains unclear. We performed immunohistochemical analysis of 99 UTUC samples to evaluate the expression status of Trop-2 in patients with UTUC and analyze its association with clinical outcomes. Trop-2 was positive in 94 of the 99 UTUC samples, and high Trop-2 expression was associated with favorable progression-free survival (PFS) and cancer-specific survival (p = 0.0011, 0.0046). Multivariate analysis identified high Trop-2 expression as an independent predictor of favorable PFS (all cases, p = 0.045; high-risk group (pT3≤ or presence of lymphovascular invasion or lymph node metastasis), p = 0.014). Gene expression analysis using RNA sequencing data from 72 UTUC samples demonstrated the association between high TACSTD2 expression and favorable PFS (all cases, p = 0.069; high-risk group, p = 0.029). In conclusion, we demonstrated that Trop-2 is widely expressed in UTUC. Although high Trop-2 expression was a favorable prognostic factor in UTUC, its widespread expression suggests that sacituzumab govitecan may be effective for a wide range of UTUC.
Collapse
|
16
|
Mutlu L, Tymon-Rosario J, Harold J, Menderes G. Targeted treatment options for the management of metastatic/persistent and recurrent cervical cancer. Expert Rev Anticancer Ther 2022; 22:633-645. [PMID: 35533682 DOI: 10.1080/14737140.2022.2075348] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Cervical cancer is the overall fourth most common malignancy and the fourth most common cause of cancer related deaths worldwide. Despite vaccination and screening programs, many women continue to present with advanced stage cervical cancer, wherein the treatment options have been limited. AREAS COVERED In this review, immunotherapy and the potential targeted therapies that have demonstrated promise in the treatment of persistent, recurrent, and metastatic cervical cancer are discussed. EXPERT OPINION Our global goal in the gynecologic oncology community is to eliminate cervical cancer, by increasing the uptake of preventive vaccination and screening programs. For unfortunate patients who present with metastatic, persistent, and recurrent cervical cancer, pembrolizumab with chemotherapy, with or without bevacizumab is the new first line therapy for PD-L1 positive patients. For this patient population as a second line therapy, tisotumab vedotin (i.e. ADC) has shown significant efficacy in Phase II trials, leading to FDA approval. Combination regimens inclusive of immune checkpoint inhibitors, DNA damage repair inhibitors, antibody drug conjugates are potential breakthrough treatment strategies and are currently being investigated.
Collapse
Affiliation(s)
- Levent Mutlu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| | - Joan Tymon-Rosario
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| | - Justin Harold
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| | - Gulden Menderes
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| |
Collapse
|
17
|
Topoisomerase I inhibitors: Challenges, progress and the road ahead. Eur J Med Chem 2022; 236:114304. [DOI: 10.1016/j.ejmech.2022.114304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/17/2022]
|
18
|
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: 13.0] [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
|
19
|
Pavone G, Motta L, Martorana F, Motta G, Vigneri P. A New Kid on the Block: Sacituzumab Govitecan for the Treatment of Breast Cancer and Other Solid Tumors. Molecules 2021; 26:molecules26237294. [PMID: 34885875 PMCID: PMC8659286 DOI: 10.3390/molecules26237294] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Human trophoblast cell-surface antigen-2 (Trop-2) is a membrane glycoprotein involved in cell proliferation and motility, frequently overexpressed in epithelial tumors. Thus, it represents an attractive target for anticancer therapies. Sacituzumab govitecan (SG) is a third-generation antibody-drug conjugate, consisting of an anti-Trop-2 monoclonal antibody (hRS7), a hydrolyzable linker, and a cytotoxin (SN38), which inhibits topoisomerase 1. Specific pharmacological features, such as the high antibody to payload ratio, the ultra-toxic nature of SN38, and the capacity to kill surrounding tumor cells (the bystander effect), make SG a very promising drug for cancer treatment. Indeed, unprecedented results have been observed with SG in patients with heavily pretreated advanced triple-negative breast cancer and urothelial carcinomas, and the drug has already received approval for these indications. These results are coupled with a manageable toxicity profile, with neutropenia and diarrhea as the most frequent adverse events, mainly of grades 1-2. While several trials are exploring SG activity in different tumor types and settings, potential biomarkers of response are under investigation. Among these, Trop-2 overexpression and the presence of BRCA1/2 mutations seem to be the most promising. We review the available literature concerning SG, with a focus on its toxicity spectrum and possible biomarkers of its response.
Collapse
Affiliation(s)
- Giuliana Pavone
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Medical Oncology Unit, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy
| | - Lucia Motta
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Medical Oncology Unit, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy
- Correspondence: ; Tel.: +39-095-3781959
| | - Federica Martorana
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Gianmarco Motta
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Medical Oncology Unit, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy
| | - Paolo Vigneri
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Medical Oncology Unit, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
20
|
Martín-Sabroso C, Lozza I, Torres-Suárez AI, Fraguas-Sánchez AI. Antibody-Antineoplastic Conjugates in Gynecological Malignancies: Current Status and Future Perspectives. Pharmaceutics 2021; 13:1705. [PMID: 34683998 PMCID: PMC8541375 DOI: 10.3390/pharmaceutics13101705] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/18/2022] Open
Abstract
In the last decade, antibody-drug conjugates (ADCs), normally formed by a humanized antibody and a small drug via a chemical cleavable or non-cleavable linker, have emerged as a potential treatment strategy in cancer disease. They allow to get a selective delivery of the chemotherapeutic agents at the tumor level, and, consequently, to improve the antitumor efficacy and, especially to decrease chemotherapy-related toxicity. Currently, nine antibody-drug conjugate-based formulations have been already approved and more than 80 are under clinical trials for the treatment of several tumors, especially breast cancer, lymphomas, and multiple myeloma. To date, no ADCs have been approved for the treatment of gynecological formulations, but many formulations have been developed and have reached the clinical stage, especially for the treatment of ovarian cancer, an aggressive disease with a low five-year survival rate. This manuscript analyzes the ADCs formulations that are under clinical research in the treatment of gynecological carcinomas, specifically ovarian, endometrial, and cervical tumors.
Collapse
Affiliation(s)
- Cristina Martín-Sabroso
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (C.M.-S.); (I.L.); (A.I.T.-S.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Irene Lozza
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (C.M.-S.); (I.L.); (A.I.T.-S.)
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (C.M.-S.); (I.L.); (A.I.T.-S.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Ana Isabel Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (C.M.-S.); (I.L.); (A.I.T.-S.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
21
|
Mauricio D, Zeybek B, Tymon-Rosario J, Harold J, Santin AD. Immunotherapy in Cervical Cancer. Curr Oncol Rep 2021; 23:61. [PMID: 33852056 DOI: 10.1007/s11912-021-01052-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW This review aims to summarize the current immunotherapy studies and the potential targeted therapies showing promise in the treatment of cervical cancer. RECENT FINDINGS There are promising ongoing monotherapy and combination therapy trials using different immune checkpoint inhibitors, poly adenosine diphosphate ribose polymerase inhibitors, tumor angiogenesis inhibitors (i.e., bevacizumab), antibody-drug conjugates, therapeutic vaccines, and tumor-infiltrating T lymphocytes (adoptive immunotherapy). Some of these novel modalities are also being evaluated in combination with standard platinum-based chemotherapy regimen. At this time, pembrolizumab is approved for the treatment of relapsed or metastatic programmed death ligand 1 (PD-L1) positive cervical cancer after frontline chemotherapy treatment. Multiple novel therapeutic modalities are emerging as safe and effective for the treatment of cervical cancer patients. Development and participation in investigative treatments can provide benefit and improve outcomes in cervical cancer.
Collapse
Affiliation(s)
- Dennis Mauricio
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, LSOG Bld. Room 305, 333 Cedar Street, PO Box 208063, New Haven, CT, 06520-8063, USA
| | - Burak Zeybek
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, LSOG Bld. Room 305, 333 Cedar Street, PO Box 208063, New Haven, CT, 06520-8063, USA
| | - Joan Tymon-Rosario
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, LSOG Bld. Room 305, 333 Cedar Street, PO Box 208063, New Haven, CT, 06520-8063, USA
| | - Justin Harold
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, LSOG Bld. Room 305, 333 Cedar Street, PO Box 208063, New Haven, CT, 06520-8063, USA
| | - Alessandro D Santin
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, LSOG Bld. Room 305, 333 Cedar Street, PO Box 208063, New Haven, CT, 06520-8063, USA.
| |
Collapse
|
22
|
Shen M, Liu S, Stoyanova T. The role of Trop2 in prostate cancer: an oncogene, biomarker, and therapeutic target. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2021; 9:73-87. [PMID: 33816696 PMCID: PMC8012837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/24/2020] [Indexed: 06/12/2023]
Abstract
Prostate cancer remains the second leading cause of cancer-associated deaths amongst American men. Trop2, a cell surface glycoprotein, correlates with poor clinical outcome and is highly expressed in metastatic, treatment-resistant prostate cancer. High levels of Trop2 are prognostic for biochemical recurrence. Trop2 regulates tumor growth and metastatic ability of prostate cancer. Moreover, overexpression of Trop2 drives the transdifferentiation to neuroendocrine phenotype in prostate cancer. In addition, Trop2 is overexpressed across epithelial cancers and has emerged as a promising therapeutic target in various solid epithelial cancers. The FDA (Food and Drug Administration) recently approved the use of a Trop2-targeting ADC (antibody-drug conjugate), Sacituzumab Govitecan (IMMU-132), for metastatic, triple-negative breast cancer with at least two prior therapies. Here, we review the role of Trop2 in prostate tumorigenesis and its potential as a promising biomarker and therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Michelle Shen
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University USA
| | - Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University USA
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University USA
| |
Collapse
|
23
|
Abstract
Sacituzumab govitecan (sacituzumab govitecan-hziy; Trodelvy™) is a Trop-2-directed antibody conjugated to a topoisomerase I inhibitor (SN-38) that is being developed by Immunomedics for the treatment of solid tumours, including breast cancer. In April 2020, sacituzumab govitecan received accelerated approval in the USA for the treatment of adult patients with metastatic triple-negative breast cancer (mTNBC) who have received at least two prior therapies for metastatic disease. Sacituzumab govitecan is undergoing phase III development for breast cancer in the USA and EU, and phase II development for urothelial cancer. It is also being explored for brain metastases, glioblastoma, endometrial cancer and prostate cancer. This article summarizes the milestones in the development of sacituzumab govitecan leading to this first approval for mTNBC.
Collapse
|
24
|
Tymon-Rosario J, Zeybek B, Santin AD. Novel antibody-drug conjugates: current and future roles in gynecologic oncology. Curr Opin Obstet Gynecol 2021; 33:26-33. [PMID: 32618744 PMCID: PMC8253558 DOI: 10.1097/gco.0000000000000642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) represent a new class of drugs that combine a surface receptor-targeting antibody linked to a cytotoxic molecule. This review summarizes the current literature demonstrating their tremendous promise as therapeutic agents in the treatment of aggressive gynecologic malignancies. RECENT FINDINGS Several antigens have proven to be differentially overexpressed in a variety of gynecologic tumors when compared with normal surrounding tissue and serve as novel targets for ADC therapy. In the last few years HER2/neu, folic acid-alpha (FRα) and Trop-2 overexpression have been exploited as excellent targets by novel ADCs such as Trastuzumab emtansine (T-DM1), SYD985, IMGN853 (Mirvetuximab soravtansine) and Sacituzumab govitecan (SG, IMMU-132) in multiple tumors including ovarian, endometrial and cervical cancers. Although the selectivity of ADCs with noncleavable linkers (i.e. T-DM1) has shown negligible effect on surrounding antigen negative cells, those ADCs with cleavable linkers (i.e. SYD985, IMGN853 and SG) may kill both antigen-positive target cells and surrounding antigen-negative cells via the bystander effect. SUMMARY Preclinical data strongly supports these ADCs and ongoing clinical trials will shed further light into the potential of making these drugs part of current standard practice and providing our patients with a higher level of personalized cancer care.
Collapse
Affiliation(s)
- Joan Tymon-Rosario
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | |
Collapse
|
25
|
DeLucia DC, Cardillo TM, Ang L, Labrecque MP, Zhang A, Hopkins JE, De Sarkar N, Coleman I, da Costa RMG, Corey E, True LD, Haffner MC, Schweizer MT, Morrissey C, Nelson PS, Lee JK. Regulation of CEACAM5 and Therapeutic Efficacy of an Anti-CEACAM5-SN38 Antibody-drug Conjugate in Neuroendocrine Prostate Cancer. Clin Cancer Res 2020; 27:759-774. [PMID: 33199493 DOI: 10.1158/1078-0432.ccr-20-3396] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Neuroendocrine prostate cancer (NEPC) is an aggressive form of castration-resistant prostate cancer (CRPC) for which effective therapies are lacking. We previously identified carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) as a promising NEPC cell surface antigen. Here we investigated the scope of CEACAM5 expression in end-stage prostate cancer, the basis for CEACAM5 enrichment in NEPC, and the therapeutic potential of the CEACAM5 antibody-drug conjugate labetuzumab govitecan in prostate cancer. EXPERIMENTAL DESIGN The expression of CEACAM5 and other clinically relevant antigens was characterized by multiplex immunofluorescence of a tissue microarray comprising metastatic tumors from 34 lethal metastatic CRPC (mCRPC) cases. A genetically defined neuroendocrine transdifferentiation assay of prostate cancer was developed to evaluate mechanisms of CEACAM5 regulation in NEPC. The specificity and efficacy of labetuzumab govitecan was determined in CEACAM5+ prostate cancer cell lines and patient-derived xenografts models. RESULTS CEACAM5 expression was enriched in NEPC compared with other mCRPC subtypes and minimally overlapped with prostate-specific membrane antigen, prostate stem cell antigen, and trophoblast cell surface antigen 2 expression. We focused on a correlation between the expression of the pioneer transcription factor ASCL1 and CEACAM5 to determine that ASCL1 can drive neuroendocrine reprogramming of prostate cancer which is associated with increased chromatin accessibility of the CEACAM5 core promoter and CEACAM5 expression. Labetuzumab govitecan induced DNA damage in CEACAM5+ prostate cancer cell lines and marked antitumor responses in CEACAM5+ CRPC xenograft models including chemotherapy-resistant NEPC. CONCLUSIONS Our findings provide insights into the scope and regulation of CEACAM5 expression in prostate cancer and strong support for clinical studies of labetuzumab govitecan for NEPC.
Collapse
Affiliation(s)
- Diana C DeLucia
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Lisa Ang
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark P Labrecque
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Ailin Zhang
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - James E Hopkins
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Navonil De Sarkar
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ilsa Coleman
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rui M Gil da Costa
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Lawrence D True
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Michael C Haffner
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Michael T Schweizer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Colm Morrissey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Urology, University of Washington School of Medicine, Seattle, Washington.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - John K Lee
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
26
|
Lenárt S, Lenárt P, Šmarda J, Remšík J, Souček K, Beneš P. Trop2: Jack of All Trades, Master of None. Cancers (Basel) 2020; 12:E3328. [PMID: 33187148 PMCID: PMC7696911 DOI: 10.3390/cancers12113328] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Trophoblast cell surface antigen 2 (Trop2) is a widely expressed glycoprotein and an epithelial cell adhesion molecule (EpCAM) family member. Although initially identified as a transmembrane protein, other subcellular localizations and processed forms were described. Its congenital mutations cause a gelatinous drop-like corneal dystrophy, a disease characterized by loss of barrier function in corneal epithelial cells. Trop2 is considered a stem cell marker and its expression associates with regenerative capacity in various tissues. Trop2 overexpression was described in tumors of different origins; however, functional studies revealed both oncogenic and tumor suppressor roles. Nevertheless, therapeutic potential of Trop2 was recognized and clinical studies with drug-antibody conjugates have been initiated in various cancer types. One of these agents, sacituzumab govitecan, has been recently granted an accelerated approval for therapy of metastatic triple-negative breast cancer. In this article, we review the current knowledge about the yet controversial function of Trop2 in homeostasis and pathology.
Collapse
Affiliation(s)
- Sára Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
| | - Peter Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Jan Šmarda
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
| | - Ján Remšík
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Karel Souček
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 65 Brno, Czech Republic
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Petr Beneš
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| |
Collapse
|
27
|
Theocharopoulos C, Lialios PP, Gogas H, Ziogas DC. An overview of antibody-drug conjugates in oncological practice. Ther Adv Med Oncol 2020; 12:1758835920962997. [PMID: 33088347 PMCID: PMC7543133 DOI: 10.1177/1758835920962997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/07/2020] [Indexed: 01/11/2023] Open
Abstract
Antibody–drug conjugates (ADCs) are designed to manipulate the toxic efficacy of
specific chemotherapeutic compounds, employing the high affinity of
antibody-mediated delivery so as to drive them selectively to target cancer
cells. These immunoconjugates encompass the general tendency towards precision
medicine and avert the systemic toxicities of conventional chemotherapy,
accomplishing an improved therapeutic index. Cumulative experience acquired from
first-generation ADCs offers new perspectives to these promising therapeutic
modalities for various hematological and solid cancers and propels their
clinical development in a faster-than-ever pace, as indicated by the approval of
four novel ADCs during the last year. This paper aims to provide an up-to-date
overview of the eight ADCs approved by the US Food and Drug Administration and
their current indications in oncological practice. Starting from their
bio-pharmaceutical background, we track their clinical evolution, with an
emphasis on the pivotal trials that led to their commercial release. Late-stage
studies examining these eight ADCs in other-than-approved settings as well as
the investigation of potential new candidates are also reviewed. In the close
future, more data are expected to expand ADCs’ oncological utility and to
further reshape their role in cancer therapeutics.
Collapse
Affiliation(s)
- Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Panagiotis-Petros Lialios
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Agiou Thoma 17, Athens, 115 27, Greece
| |
Collapse
|
28
|
Goldenberg DM, Sharkey RM. Sacituzumab govitecan, a novel, third-generation, antibody-drug conjugate (ADC) for cancer therapy. Expert Opin Biol Ther 2020; 20:871-885. [PMID: 32301634 DOI: 10.1080/14712598.2020.1757067] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION We describe a new, third-generation of antibody-drug conjugates (ADCs) having a high drug payload against topoisomerase I, important for DNA function, and targeting selective tumor antigens, predominantly TROP-2. AREAS COVERED The historical development of ADCs is reviewed before presenting the current line of improved, third-generation ADCs targeting topoisomerase I, thus affecting DNA and causing double-stranded DNA breaks. Emphasis is given to explaining why sacituzumab govitecan represents a paradigm change in ADCs by achieving a high therapeutic index due to its novel target, TROP-2, an internalizing antigen/antibody, proprietary linker chemistry, and high drug payload, resulting in a high tumor concentration of the drug given in repeated doses with acceptable tolerability, particularly evidencing a lower percentage of 'late' diarrhea than its prodrug, irinotecan. PubMed was used for the primary search conducted. EXPERT OPINION The properties and clinical results of third-generation ADCs, based on sacituzumab govitecan, are discussed, including prospects for future applications, particularly combination therapies with PARP inhibitors and immune checkpoint inhibitors. Since one topoisomerase I ADC has just received regulatory approval for HER2+ breast cancer, and sacituzumab govitecan is under FDA review for accelerated approval in the therapy of triple-negative breast cancer, the prospects for these novel ADCs are discussed.
Collapse
Affiliation(s)
| | - Robert M Sharkey
- Center for Molecular Medicine and Immunology , Mendham, New Jersey, USA
| |
Collapse
|