1
|
Matoba Y, Devins KM, Milane L, Manning WB, Mazina V, Yeku OO, Rueda BR. High-Grade Endometrial Cancer: Molecular Subtypes, Current Challenges, and Treatment Options. Reprod Sci 2024; 31:2541-2559. [PMID: 38658487 DOI: 10.1007/s43032-024-01544-5] [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: 10/02/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
Although many recent advancements have been made in women's health, perhaps one of the most neglected areas of research is the diagnosis and treatment of high-grade endometrial cancer (EnCa). The molecular classification of EnCa in concert with histology was a major step forward. The integration of profiling for mismatch repair deficiency and Human Epidermal Growth Factor 2 (HER2) overexpression, can further inform treatment options, especially for drug resistant recurrent disease. Recent early phase trials suggest that regardless of subtype, combination therapy with agents that have distinct mechanisms of action is a fruitful approach to the treatment of high-grade EnCa. Unfortunately, although the importance of diagnosis and treatment of high-grade EnCa is well recognized, it is understudied compared to other gynecologic and breast cancers. There remains a tremendous need to couple molecular profiling and biomarker development with promising treatment options to inform new treatment strategies with higher efficacy and safety for all who suffer from high-grade recurrent EnCa.
Collapse
Affiliation(s)
- Yusuke Matoba
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 60 Blossom St, 02114, Boston, MA, USA
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, 02115, Boston, MA, USA
| | - Kyle M Devins
- Department of Pathology, Massachusetts General Hospital, 021151, Boston, MA, USA
| | - Lara Milane
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, 02115, Boston, MA, USA
| | - William B Manning
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 60 Blossom St, 02114, Boston, MA, USA
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, 02115, Boston, MA, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 02114, Boston, MA, USA
| | - Varvara Mazina
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 60 Blossom St, 02114, Boston, MA, USA
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, 02115, Boston, MA, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 02114, Boston, MA, USA
| | - Oladapo O Yeku
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 60 Blossom St, 02114, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, 55 Fruit St, 02114, Boston, MA, USA
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 60 Blossom St, 02114, Boston, MA, USA.
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, 02115, Boston, MA, USA.
| |
Collapse
|
2
|
Sarkar S, Saha SA, Swarnakar A, Chakrabarty A, Dey A, Sarkar P, Banerjee S, Mitra P. The molecular prognostic score, a classifier for risk stratification of high-grade serous ovarian cancer. J Ovarian Res 2024; 17:159. [PMID: 39095849 PMCID: PMC11296390 DOI: 10.1186/s13048-024-01482-5] [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: 05/14/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The clinicopathological parameters such as residual tumor, grade, the International Federation of Gynecology and Obstetrics (FIGO) score are often used to predict the survival of ovarian cancer patients, but the 5-year survival of high grade serous ovarian cancer (HGSOC) still remains around 30%. Hence, the relentless pursuit of enhanced prognostic tools for HGSOC, this study introduces an unprecedented gene expression-based molecular prognostic score (mPS). Derived from a novel 20-gene signature through Least Absolute Shrinkage and Selection Operator (LASSO)-Cox regression, the mPS stands out for its predictive prowess. RESULTS Validation across diverse datasets, including training and test sets (n = 491 each) and a large HGSOC patient cohort from the Ovarian Tumor Tissue Analysis (OTTA) consortium (n = 7542), consistently shows an area-under-curve (AUC) around 0.7 for predicting 5-year overall survival. The mPS's impact on prognosis resonates profoundly, yielding an adjusted hazard-ratio (HR) of 6.1 (95% CI: 3.65-10.3; p < 0.001), overshadowing conventional parameters-FIGO score, residual disease, and age. Molecular insights gleaned from mPS stratification uncover intriguing pathways, with focal-adhesion, Wnt, and Notch signaling upregulated, and antigen processing and presentation downregulated (p < 0.001) in high-risk HGSOC cohorts. CONCLUSION Positioned as a robust prognostic marker, the 20-gene signature-derived mPS emerges as a potential game-changer in clinical settings. Beyond its role in predicting overall survival, its implications extend to guiding alternative therapies, especially targeting Wnt/Notch signaling pathways and immune evasion-a promising avenue for improving outcomes in high-risk HGSOC patients.
Collapse
Affiliation(s)
- Siddik Sarkar
- Cancer Biology & Inflammatory Disorder, Translational Research Unit of Excellence (TRUE), CSIR-Indian Institute of Chemical Biology, Kolkata, WB, 700032, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP, 201002, India.
| | - Sarbar Ali Saha
- Cancer Biology & Inflammatory Disorder, Translational Research Unit of Excellence (TRUE), CSIR-Indian Institute of Chemical Biology, Kolkata, WB, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP, 201002, India
| | - Abhishek Swarnakar
- Cancer Biology & Inflammatory Disorder, Translational Research Unit of Excellence (TRUE), CSIR-Indian Institute of Chemical Biology, Kolkata, WB, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP, 201002, India
| | - Arnab Chakrabarty
- Cancer Biology & Inflammatory Disorder, Translational Research Unit of Excellence (TRUE), CSIR-Indian Institute of Chemical Biology, Kolkata, WB, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP, 201002, India
| | - Avipsa Dey
- Cancer Biology & Inflammatory Disorder, Translational Research Unit of Excellence (TRUE), CSIR-Indian Institute of Chemical Biology, Kolkata, WB, 700032, India
| | - Poulomi Sarkar
- Cancer Biology & Inflammatory Disorder, Translational Research Unit of Excellence (TRUE), CSIR-Indian Institute of Chemical Biology, Kolkata, WB, 700032, India
| | - Sarthak Banerjee
- Cancer Biology & Inflammatory Disorder, Translational Research Unit of Excellence (TRUE), CSIR-Indian Institute of Chemical Biology, Kolkata, WB, 700032, India
| | - Pralay Mitra
- Computer Science and Engineering, Indian Institute of Technology Kharagpur, Kharagpur, WB, 721302, India
| |
Collapse
|
3
|
Fernandes I, Chehade R, MacKay H. PARP inhibitors in non-ovarian gynecologic cancers. Ther Adv Med Oncol 2024; 16:17588359241255174. [PMID: 38882441 PMCID: PMC11179472 DOI: 10.1177/17588359241255174] [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: 01/21/2024] [Accepted: 04/25/2024] [Indexed: 06/18/2024] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) have transformed the treatment of ovarian cancer, particularly benefiting patients whose tumors harbor genomic events that result in impaired homologous recombination (HR) repair. The use of PARPi over recent years has expanded to include subpopulations of patients with breast, pancreatic, and prostate cancers. Their potential to benefit patients with non-ovarian gynecologic cancers is being recognized. This review examines the underlying biological rationale for exploring PARPi in non-ovarian gynecologic cancers. We consider the clinical data and place this in the context of the current treatment landscape. We review the development of PARPi strategies for treating patients with endometrial, cervical, uterine leiomyosarcoma, and vulvar cancers. Furthermore, we discuss future directions and the importance of understanding HR deficiency in the context of each cancer type.
Collapse
Affiliation(s)
| | - Rania Chehade
- Sunnybrook Odette Cancer Centre, Toronto, ON, Canada
| | - Helen MacKay
- Sunnybrook Odette Cancer Centre, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
4
|
Freyer G, Floquet A, Tredan O, Carrot A, Langlois-Jacques C, Lopez J, Selle F, Abdeddaim C, Leary A, Dubot-Poitelon C, Fabbro M, Gladieff L, Lamuraglia M. Bevacizumab, olaparib, and durvalumab in patients with relapsed ovarian cancer: a phase II clinical trial from the GINECO group. Nat Commun 2024; 15:1985. [PMID: 38443333 PMCID: PMC10914754 DOI: 10.1038/s41467-024-45974-w] [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: 12/29/2022] [Accepted: 02/08/2024] [Indexed: 03/07/2024] Open
Abstract
Most patients with advanced ovarian cancer (AOC) ultimately relapse after platinum-based chemotherapy. Combining bevacizumab, olaparib, and durvalumab likely drives synergistic activity. This open-label phase 2 study (NCT04015739) aimed to assess activity and safety of this triple combination in female patients with relapsed high-grade AOC following prior platinum-based therapy. Patients were treated with olaparib (300 mg orally, twice daily), the bevacizumab biosimilar FKB238 (15 mg/kg intravenously, once-every-3-weeks), and durvalumab (1.12 g intravenously, once-every-3-weeks) in nine French centers. The primary endpoint was the non-progression rate at 3 months for platinum-resistant relapse or 6 months for platinum-sensitive relapse per RECIST 1.1 and irRECIST. Secondary endpoints were CA-125 decline with CA-125 ELIMination rate constant K (KELIM-B) per CA-125 longitudinal kinetics over 100 days, progression free survival and overall survival, tumor response, and safety. Non-progression rates were 69.8% (90%CI 55.9%-80.0%) at 3 months for platinum-resistant relapse patients (N = 41), meeting the prespecified endpoint, and 43.8% (90%CI 29.0%-57.4%) at 6 months for platinum-sensitive relapse (N = 33), not meeting the prespecified endpoint. Median progression-free survival was 4.1 months (95%CI 3.5-5.9) and 4.9 months (95%CI 2.9-7.0) respectively. Favorable KELIM-B was associated with better survival. No toxic deaths or major safety signals were observed. Here we show that further investigation of this triple combination may be considered in AOC patients with platinum-resistant relapse.
Collapse
Affiliation(s)
- Gilles Freyer
- Department of Medical Oncology, Lyon 1 University, Lyon, France.
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France.
- Institut de Cancérologie des HCL, Lyon, France.
| | - Anne Floquet
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Department of Medical Oncology - Gynecological Tumors, Institut Bergonié, Bordeaux, France
| | - Olivier Tredan
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Aurore Carrot
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- EMR 3738, UFR Lyon-Sud, Université Lyon1, Lyon, France
| | - Carole Langlois-Jacques
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Biostatistics and Bioinformatics Department, Hospices Civils de Lyon, Lyon, France
| | - Jonathan Lopez
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Department of Biochemistry and Molecular Biology, Hospices Civils de Lyon, Lyon, France
| | - Frédéric Selle
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Department of Medical Oncology, Groupe Hospitalier Diaconesses Croix Saint-Simon, Paris, France
| | - Cyril Abdeddaim
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Gynecologic Oncology Department, Centre Oscar Lambret, Lille, France
| | - Alexandra Leary
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Oncology Department, Institut Gustave Roussy, Villejuif, France
| | - Coraline Dubot-Poitelon
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Medical Oncology, Institut Curie Saint Cloud, Paris, France
| | - Michel Fabbro
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - Laurence Gladieff
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Medical Oncology, Institut Claudius Regaud IUCT-Oncopole, Toulouse, France
| | - Michele Lamuraglia
- GINECO (Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'Ovaire, Paris, France
- Medical Oncology, Institut de Cancérologie du CHUSE, Saint-Etienne, France
| |
Collapse
|
5
|
Xia D, Xu X, Du P. Comments on "Randomized, two-arm, non-comparative phase 2 study of olaparib plus cediranib or durvalumab in HRR-mutated, platinum-resistant ovarian cancer: A substudy of KGOG 3045". Int J Cancer 2024; 154:762-763. [PMID: 37907806 DOI: 10.1002/ijc.34785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023]
Affiliation(s)
- Dongyue Xia
- Department of Gynaecology and Obstetrics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Xueyuan Xu
- Department of Gynaecology and Obstetrics, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, China
- South China Normal University, Guangzhou, China
| | - Pei Du
- Department of Gynaecology and Obstetrics, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Skouteris N, Papageorgiou G. PARP Inhibitors in Colorectal Malignancies: A 2023 Update. Rev Recent Clin Trials 2024; 19:101-108. [PMID: 38058097 DOI: 10.2174/0115748871260815231116060817] [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: 06/11/2023] [Revised: 09/06/2023] [Accepted: 09/28/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Colorectal carcinoma (CRC) is one of the most common malignancies in the Western world, and metastatic disease is associated with a dismal prognosis. Poly-ADpribose polymerase (PARP) inhibitors gain increasing attention in the field of medical oncology, as they lead to synthetic lethality in malignancies with preexisting alterations in the DNA damage repair (DDR) pathway. As those alterations are frequently seen in CRC, a targeted approach through PARP inhibitors is expected to benefit these patients, both alone and in combination with other agents like chemotherapy, immunotherapy, antiangiogenics, and radiation. OBJECTIVE This review article aims to better clarify the role of PARP inhibitors as a treatment option in patients with metastatic CRC with alterations in the DDR pathway. METHODS We used the PubMed database to retrieve journal articles and the inclusion criteria were all human studies that illustrated the effective role of PARP inhibitors in patients with metastatic CRC with homologous repair deficiency (HRD) and the correct line of therapy. RESULTS Current evidence supports the utilization of PARP inhibitors in CRC subgroups, as monotherapy and in combination with other agents. Up to now, data are insufficient to support a formal indication, and further research is needed. CONCLUSION Efforts to precisely define the homologous repair deficiency (HRD) in CRC - and eventually the subgroup of patients that are expected to benefit the most - are also underway.
Collapse
Affiliation(s)
- Nikolaos Skouteris
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, "Metaxa" Cancer Hospital, 51 Botassi Street, 18537 Piraeus, Greece
| | | |
Collapse
|
7
|
Fan Z, Han D, Fan X, Zhao L. Ovarian cancer treatment and natural killer cell-based immunotherapy. Front Immunol 2023; 14:1308143. [PMID: 38187402 PMCID: PMC10768003 DOI: 10.3389/fimmu.2023.1308143] [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: 10/05/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Background Ovarian cancer (OC) is one of the malignant tumors that poses a serious threat to women's health. Natural killer (NK) cells are an integral part of the immune system and have the ability to kill tumor cells directly or participate indirectly in the anti-tumor immune response. In recent years, NK cell-based immunotherapy for OC has shown remarkable potential. However, its mechanisms and effects remain unclear when compared to standard treatment. Methods To explore the value of NK cell-based immunotherapy in the treatment of OC, we conducted a literature review. In comparison to standard treatment, our focus was primarily on the current anti-tumor mechanisms, the clinical effect of NK cells against OC, factors affecting the structure and function of NK cells, and strategies to enhance the effectiveness of NK cells. Results We found that NK cells exert their therapeutic effects in OC through mechanisms such as antibody-dependent cell cytotoxicity, perforin release, and granule enzyme secretion. They also secrete IFN-γ and TNF-α or engage in Fas/FasL and TRAIL/TRAILR pathways, mediating the death of OC cells. In clinical trials, the majority of patients experienced disease stability with mild side effects after receiving NK cell-based immunotherapy, but there is still a lack of high-quality research evidence regarding its clinical effectiveness. OC and prior experience with standard treatments have an effect on NK cells, and it may be considered to maximize NK cell effects through the modulation of the tumor microenvironment or combination with other therapies. Conclusions In this review, we have summarized the current evidence of NK cell applications in the treatment of OC. Furthermore, factors and strategies that influence and enhance the role of NK cell immunotherapy are discussed.
Collapse
Affiliation(s)
- Zhongru Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Dongyu Han
- Department of Obstetrics and Gynecology, Suzhou Hospital, Affiliated Hospital of Meddical School, Nanjing University, Suzhou, China
| | - Xin Fan
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Zhao
- Department of Obstetrics and Gynecology, Suzhou Hospital, Affiliated Hospital of Meddical School, Nanjing University, Suzhou, China
| |
Collapse
|
8
|
Wang J, Ford JC, Mitra AK. Defining the Role of Metastasis-Initiating Cells in Promoting Carcinogenesis in Ovarian Cancer. BIOLOGY 2023; 12:1492. [PMID: 38132318 PMCID: PMC10740540 DOI: 10.3390/biology12121492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Ovarian cancer is the deadliest gynecological malignancy with a high prevalence of transcoelomic metastasis. Metastasis is a multi-step process and only a small percentage of cancer cells, metastasis-initiating cells (MICs), have the capacity to finally establish metastatic lesions. These MICs maintain a certain level of stemness that allows them to differentiate into other cell types with distinct transcriptomic profiles and swiftly adapt to external stresses. Furthermore, they can coordinate with the microenvironment, through reciprocal interactions, to invade and establish metastases. Therefore, identifying, characterizing, and targeting MICs is a promising strategy to counter the spread of ovarian cancer. In this review, we provided an overview of OC MICs in the context of characterization, identification through cell surface markers, and their interactions with the metastatic niche to promote metastatic colonization.
Collapse
Affiliation(s)
- Ji Wang
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN 47405, USA; (J.W.); (J.C.F.)
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
| | - James C. Ford
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN 47405, USA; (J.W.); (J.C.F.)
| | - Anirban K. Mitra
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN 47405, USA; (J.W.); (J.C.F.)
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
Shan C, Wang Y, Li Y, Yang S, Sheng W, Liu X. The triple-drug combination DBDx enhances the antitumor efficacy of PD-1 antibody associated with Treg modulation. J Cancer Res Ther 2023; 19:1603-1609. [PMID: 38156928 DOI: 10.4103/jcrt.jcrt_350_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/03/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE This study investigated the antitumor efficacy of programmed cell death protein-1 (PD-1) antibody and DBDx, a triple-drug combination of dipyridamole, bestatin, and dexamethasone, and their related immunomodulation. MATERIALS AND METHODS Mouse melanoma B16, mouse Lewis lung carcinoma, and mouse breast carcinoma 4T1 were used for evaluating the in vivo therapeutic efficacy of DBDx, PD-1 antibody, and their combination. The peripheral blood and tumor tissues of 4T1 tumor-bearing mice were collected to analyze regulatory T cells and measured using flow cytometry. RESULTS The combination of PD-1 antibody and DBDx enhanced the therapeutic efficacy against B16 melanoma. The suppression of tumor growth by PD-1 antibody and DBDx was more significant than that by anti-PD-1 monotherapy. The tumor growth inhibition rates of PD-1 antibody, DBDx, and their combination were 54.0%, 72.4%, and 83.1%, respectively, suggesting a synergistic effect as determined by the coefficient of drug interaction. No significant changes were found in the body weights in all the above groups, indicating that the treated mice tolerated the applied drug doses. Similarly, enhanced therapeutic efficacy of the PD-1 antibody and DBDx combination was observed in murine Lewis lung carcinoma and 4T1 breast cancer models. In 4T1 breast cancer-bearing mice, the immunotherapy-related changes in lymphocytes in peripheral blood and tumor microenvironment were evaluated with flow cytometry. Compared with anti-PD-1 monotherapy, peripheral blood and tumor-infiltrating lymphocytes were found a lower ratio of regulatory T cell (Treg) subset cells and a higher ratio of CD8+/Treg cells. CONCLUSIONS The combination of PD-1 antibody and DBDx could achieve enhanced therapeutic antitumor efficacy than anti-PD-1 monotherapy, suggesting potential for using the triple-drug combination DBDx in cancer immunotherapy.
Collapse
Affiliation(s)
- Chuankun Shan
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, P.R. China
- Jiangsu Alphamab Biopharmaceuticals Co., Ltd, Suzhou, P.R. China
| | - Yuexuan Wang
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, P.R. China
- Zibo Central Hospital, Zibo, P.R. China
| | - Yi Li
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, P.R. China
| | - Siqi Yang
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, P.R. China
- Qujing Center for Disease Control and Prevention, Qujing, P.R. China
| | - Weijin Sheng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, P.R. China
| | - Xiujun Liu
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, P.R. China
| |
Collapse
|
10
|
Fanale D, Corsini LR, Pedone E, Randazzo U, Fiorino A, Di Piazza M, Brando C, Magrin L, Contino S, Piraino P, Bazan Russo TD, Cipolla C, Russo A, Bazan V. Potential agnostic role of BRCA alterations in patients with several solid tumors: One for all, all for one? Crit Rev Oncol Hematol 2023; 190:104086. [PMID: 37536445 DOI: 10.1016/j.critrevonc.2023.104086] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023] Open
Abstract
Germline BRCA1/2 alterations in the Homologous Recombination (HR) pathway are considered as main susceptibility biomarkers to Hereditary Breast and Ovarian Cancers (HBOC). The modern molecular biology technologies allowed to characterize germline and somatic BRCA1/2 alterations in several malignancies, broadening the landscape of BRCA1/2-alterated tumors. In the last years, BRCA genetic testing, beyond the preventive value, also assumed a predictive and prognostic significance for patient management. The approval of molecules with agnostic indication is leading to a new clinical model, defined "mutational". Among these drugs, the Poly (ADP)-Ribose Polymerase inhibitors (PARPi) for BRCA1/2-deficient tumors were widely studied leading to increasing therapeutic implications. In this Review we provided an overview of the main clinical studies describing the association between BRCA-mutated tumors and PARPi response, focusing on the controversial evidence about the potential agnostic indication based on BRCA1/2 alterations in several solid tumors.
Collapse
Affiliation(s)
- Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Ugo Randazzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessia Fiorino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Marianna Di Piazza
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Luigi Magrin
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Silvia Contino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Paola Piraino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Calogero Cipolla
- Division of General and Oncological Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
11
|
Johnson RL, Ganesan S, Thangavelu A, Theophilou G, de Jong D, Hutson R, Nugent D, Broadhead T, Laios A, Cummings M, Orsi NM. Immune Checkpoint Inhibitors Targeting the PD-1/PD-L1 Pathway in Advanced, Recurrent Endometrial Cancer: A Scoping Review with SWOT Analysis. Cancers (Basel) 2023; 15:4632. [PMID: 37760602 PMCID: PMC10527181 DOI: 10.3390/cancers15184632] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Results of recent clinical trials using the immune check point inhibitors (ICI) pembrolizumab or dostarlimab with/without lenvatinib has led to their approval for specific molecular subgroups of advanced recurrent endometrial cancer (EC). Herein, we summarise the clinical data leading to this first tissue-agnostic approval. As this novel therapy is not yet available in the United Kingdom standard care setting, we explore the strengths, weaknesses, opportunities, and threats (SWOT) of ICI treatment in EC. Major databases were searched focusing on clinical trials using programmed cell death protein 1 (PD-1) and its ligand (PD-L1) ICI which ultimately contributed to anti-PD-1 approval in EC. We performed a data quality assessment, reviewing survival and safety analysis. We included 15 studies involving 1609 EC patients: 458 with mismatch repair deficiency (MMRd)/microsatellite instability-high (MSI-H) status and 1084 with mismatch repair proficiency/microsatellite stable (MMRp/MSS) status. Pembrolizumab/dostarlimab have been approved for MMRd ECs, with the addition of lenvatinib for MMRp cases in the recurrent setting. Future efforts will focus on the pathological assessment of biomarkers to determine molecular phenotypes that correlate with response or resistance to ICI in order to identify patients most likely to benefit from this treatment.
Collapse
Affiliation(s)
- Racheal Louise Johnson
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Subhasheenee Ganesan
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Amudha Thangavelu
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Georgios Theophilou
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Diederick de Jong
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Richard Hutson
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - David Nugent
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Timothy Broadhead
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Alexandros Laios
- Department of Gynaecological Oncology, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Michele Cummings
- Leeds Institute of Medical Research, St James’s University Hospital, The University of Leeds, Leeds LS9 7TF, UK
| | - Nicolas Michel Orsi
- Leeds Institute of Medical Research, St James’s University Hospital, The University of Leeds, Leeds LS9 7TF, UK
| |
Collapse
|
12
|
Zhao L, Chen X, Wu H, He Q, Ding L, Yang B. Strategies to synergize PD-1/PD-L1 targeted cancer immunotherapies to enhance antitumor responses in ovarian cancer. Biochem Pharmacol 2023; 215:115724. [PMID: 37524205 DOI: 10.1016/j.bcp.2023.115724] [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: 04/11/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Anti-programmed cell death 1/programmed cell death ligand 1 (anti-PD-1/PD-L1) antibodies have developed rapidly but exhibited modest activity in ovarian cancer (OC), achieving a clinical response rate ranging from 5.9% to 19%. Current evidence indicate that the establishment of an integrated cancer-immunity cycle is a prerequisite for anti-PD-1/PD-L1 antibodies. Any impairment in this cycle, including lack of cancer antigens release, impaired antigen-presenting, decreased T cell priming and activation, less T cells that are trafficked or infiltrated in tumor microenvironment (TME), and low tumor recognition and killings, will lead to decreased infiltrated cytotoxic T cells to tumor bed and treatment failure. Therefore, combinatorial strategies aiming to modify cancer-immunity cycle and reprogram tumor immune microenvironment are of great interest. By far, various strategies have been studied to enhance responsiveness to PD-1/PD-L1 inhibitors in OC. Platinum-based chemotherapy increases neoantigens release; poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis) improve the function of antigen-presenting cells and promote the trafficking of T cells into tumors; epigenetic drugs help to complete the immune cycle by affecting multiple steps; immunotherapies like anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) antibodies reactivate T cells, and other treatment strategies like radiotherapy helps to increase the expression of tumor antigens. In this review, we will summarize the preclinical studies by analyzing their contribution in modifying the cancer immunity cycle and remodeling tumor environment, and we will also summarize recent progress in clinical trials and discuss some perspectives to improve these treatment strategies.
Collapse
Affiliation(s)
- Lin Zhao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xi Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
13
|
Montazeri Aliabadi H, Manda A, Sidgal R, Chung C. Targeting Breast Cancer: The Familiar, the Emerging, and the Uncharted Territories. Biomolecules 2023; 13:1306. [PMID: 37759706 PMCID: PMC10526846 DOI: 10.3390/biom13091306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer became the most diagnosed cancer in the world in 2020. Chemotherapy is still the leading clinical strategy in breast cancer treatment, followed by hormone therapy (mostly used in hormone receptor-positive types). However, with our ever-expanding knowledge of signaling pathways in cancer biology, new molecular targets are identified for potential novel molecularly targeted drugs in breast cancer treatment. While this has resulted in the approval of a few molecularly targeted drugs by the FDA (including drugs targeting immune checkpoints), a wide array of signaling pathways seem to be still underexplored. Also, while combinatorial treatments have become common practice in clinics, the majority of these approaches seem to combine molecularly targeted drugs with chemotherapeutic agents. In this manuscript, we start by analyzing the list of FDA-approved molecularly targeted drugs for breast cancer to evaluate where molecular targeting stands in breast cancer treatment today. We will then provide an overview of other options currently under clinical trial or being investigated in pre-clinical studies.
Collapse
Affiliation(s)
- Hamidreza Montazeri Aliabadi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | | | | | | |
Collapse
|
14
|
Wooten J, Mavingire N, Damar K, Loaiza-Perez A, Brantley E. Triumphs and challenges in exploiting poly(ADP-ribose) polymerase inhibition to combat triple-negative breast cancer. J Cell Physiol 2023; 238:1625-1640. [PMID: 37042191 DOI: 10.1002/jcp.31015] [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: 01/12/2023] [Accepted: 03/14/2023] [Indexed: 04/13/2023]
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) regulates a myriad of DNA repair mechanisms to preserve genomic integrity following DNA damage. PARP inhibitors (PARPi) confer synthetic lethality in malignancies with a deficiency in the homologous recombination (HR) pathway. Patients with triple-negative breast cancer (TNBC) fail to respond to most targeted therapies because their tumors lack expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Certain patients with TNBC harbor mutations in HR mediators such as breast cancer susceptibility gene 1 (BRCA1) and breast cancer susceptibility gene 2 (BRCA2), enabling them to respond to PARPi. PARPi exploits the synthetic lethality of BRCA-mutant cells. However, de novo and acquired PARPi resistance frequently ensue. In this review, we discuss the roles of PARP in mediating DNA repair processes in breast epithelial cells, mechanisms of PARPi resistance in TNBC, and recent advances in the development of agents designed to overcome PARPi resistance in TNBC.
Collapse
Affiliation(s)
- Jonathan Wooten
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Loma Linda University Health, Loma Linda, California, USA
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University Health, Loma Linda, California, USA
| | - Nicole Mavingire
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Loma Linda University Health, Loma Linda, California, USA
| | - Katherine Damar
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Loma Linda University Health, Loma Linda, California, USA
| | - Andrea Loaiza-Perez
- Facultad de Medicina, Instituto de Oncología Ángel H. Roffo (IOAHR), Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Eileen Brantley
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Loma Linda University Health, Loma Linda, California, USA
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University Health, Loma Linda, California, USA
| |
Collapse
|
15
|
Jin Z, Zhou Q, Cheng JN, Jia Q, Zhu B. Heterogeneity of the tumor immune microenvironment and clinical interventions. Front Med 2023; 17:617-648. [PMID: 37728825 DOI: 10.1007/s11684-023-1015-9] [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: 02/15/2023] [Accepted: 06/24/2023] [Indexed: 09/21/2023]
Abstract
The tumor immune microenvironment (TIME) is broadly composed of various immune cells, and its heterogeneity is characterized by both immune cells and stromal cells. During the course of tumor formation and progression and anti-tumor treatment, the composition of the TIME becomes heterogeneous. Such immunological heterogeneity is not only present between populations but also exists on temporal and spatial scales. Owing to the existence of TIME, clinical outcomes can differ when a similar treatment strategy is provided to patients. Therefore, a comprehensive assessment of TIME heterogeneity is essential for developing precise and effective therapies. Facilitated by advanced technologies, it is possible to understand the complexity and diversity of the TIME and its influence on therapy responses. In this review, we discuss the potential reasons for TIME heterogeneity and the current approaches used to explore it. We also summarize clinical intervention strategies based on associated mechanisms or targets to control immunological heterogeneity.
Collapse
Affiliation(s)
- Zheng Jin
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China
- Research Institute, GloriousMed Clinical Laboratory (Shanghai) Co. Ltd., Shanghai, 201318, China
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Qin Zhou
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jia-Nan Cheng
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| | - Qingzhu Jia
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| | - Bo Zhu
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| |
Collapse
|
16
|
Wilczyński JR, Wilczyński M, Paradowska E. "DEPHENCE" system-a novel regimen of therapy that is urgently needed in the high-grade serous ovarian cancer-a focus on anti-cancer stem cell and anti-tumor microenvironment targeted therapies. Front Oncol 2023; 13:1201497. [PMID: 37448521 PMCID: PMC10338102 DOI: 10.3389/fonc.2023.1201497] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer, especially high-grade serous type, is the most lethal gynecological malignancy. The lack of screening programs and the scarcity of symptomatology result in the late diagnosis in about 75% of affected women. Despite very demanding and aggressive surgical treatment, multiple-line chemotherapy regimens and both approved and clinically tested targeted therapies, the overall survival of patients is still unsatisfactory and disappointing. Research studies have recently brought some more understanding of the molecular diversity of the ovarian cancer, its unique intraperitoneal biology, the role of cancer stem cells, and the complexity of tumor microenvironment. There is a growing body of evidence that individualization of the treatment adjusted to the molecular and biochemical signature of the tumor as well as to the medical status of the patient should replace or supplement the foregoing therapy. In this review, we have proposed the principles of the novel regimen of the therapy that we called the "DEPHENCE" system, and we have extensively discussed the results of the studies focused on the ovarian cancer stem cells, other components of cancer metastatic niche, and, finally, clinical trials targeting these two environments. Through this, we have tried to present the evolving landscape of treatment options and put flesh on the experimental approach to attack the high-grade serous ovarian cancer multidirectionally, corresponding to the "DEPHENCE" system postulates.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, Lodz, Poland
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother's Health Center-Research Institute, Lodz, Poland
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland
| |
Collapse
|
17
|
Tu M, Xu J. Advances in immunotherapy for gynecological malignancies. Crit Rev Oncol Hematol 2023:104063. [PMID: 37385307 DOI: 10.1016/j.critrevonc.2023.104063] [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: 02/11/2023] [Revised: 05/21/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023] Open
Abstract
To date, surgery, chemotherapy and radiotherapy are mainly used to treat or remove gynecological malignancies. However, these approaches have their limitations when facing complicated female diseases such as advanced cervical and endometrial cancer (EC), chemotherapy-resistant gestational trophoblastic neoplasia and platinum-resistant ovarian cancer. Instead, immunotherapy, as an alternative, could significantly improve prognosis of those patients receiving traditional treatments, with better antitumor activities and possibly less cellular toxicities. Its' development is still not fast enough to meet the current clinical needs. More preclinical studies and larger-scale clinical trials are required. This review aims to introduce the landscape and up-to-date status of immunotherapy against gynecological malignancies, with a discussion of the challenges and future direction.
Collapse
Affiliation(s)
- Mengyan Tu
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
18
|
Yoon WH, DeFazio A, Kasherman L. Immune checkpoint inhibitors in ovarian cancer: where do we go from here? CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:358-377. [PMID: 37457131 PMCID: PMC10344730 DOI: 10.20517/cdr.2023.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/22/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023]
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynaecological malignancy, and despite advancements in therapeutics, most women unfortunately still succumb to their disease. Immunotherapies, in particular immune checkpoint inhibitors (ICI), have been therapeutically transformative in many tumour types, including gynaecological malignancies such as cervical and endometrial cancer. Unfortunately, these therapeutic successes have not been mirrored in ovarian cancer clinical studies. This review provides an overview of the ovarian tumour microenvironment (TME), particularly factors associated with survival, and explores current research into immunotherapeutic strategies in EOC, with an exploratory focus on novel therapeutics in navigating drug resistance.
Collapse
Affiliation(s)
- Won-Hee Yoon
- Department of Medical Oncology, Blacktown Cancer and Haematology Centre, Blacktown Hospital, Blacktown 2148, Australia
- Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead 2145, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead 2145, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
| | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead 2145, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
- Department of Gynecological Oncology, Westmead Hospital, Westmead 2145, Australia
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council New South Wales, Sydney 2011, Australia
| | - Lawrence Kasherman
- Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
- Department of Medical Oncology, Illawarra Cancer Care Centre, Wollongong 2500, Australia
| |
Collapse
|
19
|
Song M, Zeng X, Wu Q, Huang J, Dong J, Shao L, Sun Z, Lin Y, Chen S. Metastatic Colorectal Cancer Patient with Microsatellite Stability and Germline BRAC2 Mutation Shows a Complete Response to Olaparib in Combination with a PD-1 Inhibitor and Bevacizumab: A Case Report and Review of the Literature. Life (Basel) 2023; 13:life13051183. [PMID: 37240828 DOI: 10.3390/life13051183] [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: 04/18/2023] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Metastatic colorectal cancer (mCRC) has a poor prognosis. Combining chemotherapy with targeted therapy constitutes a basic form of mCRC treatment. Immune checkpoint inhibitors have been recommended for microsatellite instability mCRC, while most patients harboring microsatellite stability (MSS) or proficient mismatch repair (pMMR) are less responsive to immunotherapy. Combinational targeted therapy, including poly-ADP ribose polymerase (PARP) inhibitors, has been considered a promising way to reverse immunotherapy resistance; however, there is no clear and consistent conclusions can be drawn from the current research. Here, we report the case of a 59-year-old woman diagnosed with stage IVB MSS mCRC who received three courses of capecitabine/oxaliplatin chemotherapy combined with bevacizumab as a first-line treatment, resulting in an overall evaluation of stable disease (-25.7%). However, the occurrence of adverse events of intolerable grade 3 diarrhea and vomiting forced the cessation of this therapy. A germline BRCA2 mutation was found by next-generation sequencing, and the patient further received a combination of olaparib, tislelizumab, and bevacizumab. This treatment regime resulted in a complete metabolic response and a partial response (-50.9%) after 3 months of treatment. Mild asymptomatic interstitial pneumonia and manageable hematologic toxicity were two adverse events associated with this combination therapy. This study provides new insights into the combination of PARP inhibitors and immunotherapy for MSS mCRC patients carrying germline BRCA2 mutations.
Collapse
Affiliation(s)
- Minghan Song
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Xianrong Zeng
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Qian Wu
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Jie Huang
- Department of Oncology, The Fifth Affiliated Hospital of Jinan University, Heyuan 517000, China
| | - Jiayi Dong
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Lijuan Shao
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Zihao Sun
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Yiguang Lin
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangzhou Anjie Biomedical Technology Co., Ltd., Guangzhou 510530, China
| | - Size Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangdong Pharmaceutical University, Guangzhou 510080, China
| |
Collapse
|
20
|
Li T, Wang X, Qin S, Chen B, Yi M, Zhou J. Targeting PARP for the optimal immunotherapy efficiency in gynecologic malignancies. Biomed Pharmacother 2023; 162:114712. [PMID: 37075667 DOI: 10.1016/j.biopha.2023.114712] [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: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023] Open
Abstract
Gynecologic cancer, which includes ovarian, cervical, endometrial, vulvar, and vaginal cancer, is a major health concern for women all over the world. Despite the availability of various treatment options, many patients eventually progress to advanced stages and face high mortality rates. PARPi (poly (ADP-ribose) polymerase inhibitor) and immune checkpoint inhibitor (ICI) have both shown significant efficacy in the treatment of advanced and metastatic gynecologic cancer. However, both treatments have limitations, including inevitable resistance and a narrow therapeutic window, making PARPi and ICI combination therapy a promising approach to treating gynecologic malignancies. Preclinical and clinical trials have looked into the combination therapy of PARPi and ICI. PARPi improves ICI efficacy by inducing DNA damage and increasing tumor immunogenicity, resulting in a stronger immune response against cancer cells. ICI, conversly, can increase PARPi sensitivity by priming and activating immune cells, consequently prompting immune cytotoxic effect. Several clinical trials in gynecologic cancer patients have investigated the combination therapy of PARPi and ICI. When compared to monotherapy, the combination of PARPi and ICI increased progression-free survival and overall survival in ovarian cancer patients. The combination therapy has also been studied in other types of gynecologic cancer, including endometrial and cervical cancer, with promising results. Finally, the combination therapeutic strategy of PARPi and ICI is a promising approach in the treatment of gynecologic cancer, particularly advanced and metastatic stages. Preclinical studies and clinical trials have demonstrated the safety and efficacy of this combination therapy in improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Tianye Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinrun Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuang Qin
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingxin Chen
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
21
|
Jin N, Xia Y, Gao Q. Combined PARP inhibitors and small molecular inhibitors in solid tumor treatment (Review). Int J Oncol 2023; 62:28. [PMID: 36601757 PMCID: PMC9851129 DOI: 10.3892/ijo.2023.5476] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/23/2022] [Indexed: 01/05/2023] Open
Abstract
With the development of precision medicine, targeted therapy has attracted extensive attention. Poly(ADP‑ribose) polymerase inhibitors (PARPi) are critical clinical drugs designed to induce cell death and are major antitumor targeted agents. However, preclinical and clinical data have revealed the limitations of PARPi monotherapy. Therefore, their combination with other targeted drugs has become a research hotspot in tumor treatment. Recent studies have demonstrated the critical role of small molecular inhibitors in multiple haematological cancers and solid tumors via cellular signalling modulation, exhibiting potential as a combined pharmacotherapy. In the present review, studies focused on small molecular inhibitors targeting the homologous recombination pathway were summarized and clinical trials evaluating the safety and efficacy of combined treatment were discussed.
Collapse
Affiliation(s)
- Ning Jin
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Yu Xia
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Qinglei Gao
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
22
|
San-Román-Gil M, Torres-Jiménez J, Pozas J, Esteban-Villarrubia J, Albarrán-Fernández V, Álvarez-Ballesteros P, Chamorro-Pérez J, Rosero-Rodríguez D, Orejana-Martín I, Martínez-Delfrade Í, Reguera-Puertas P, Fuentes-Mateos R, Ferreiro-Monteagudo R. Current Landscape and Potential Challenges of Immune Checkpoint Inhibitors in Microsatellite Stable Metastatic Colorectal Carcinoma. Cancers (Basel) 2023; 15:863. [PMID: 36765821 PMCID: PMC9913409 DOI: 10.3390/cancers15030863] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequent cancer and the second most common cause of cancer-related death in Europe. High microsatellite instability (MSI-H) due to a deficient DNA mismatch repair (dMMR) system can be found in 5% of metastatic CRC (mCRC) and has been established as a biomarker of response to immunotherapy in these tumors. Therefore, immune checkpoint inhibitors (ICIs) in mCRC with these characteristics were evaluated with results showing remarkable response rates and durations of response. The majority of mCRC cases have high levels of DNA mismatch repair proteins (pMMR) with consequent microsatellite stability or low instability (MSS or MSI-low), associated with an inherent resistance to ICIs. This review aims to provide a comprehensive analysis of the possible approaches to overcome the mechanisms of resistance and evaluates potential biomarkers to establish the role of ICIs in pMMR/MSS/MSI-L (MSS) mCRC.
Collapse
Affiliation(s)
- María San-Román-Gil
- Medical Oncology Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | - Javier Torres-Jiménez
- Medical Oncology Department, Clínico San Carlos University Hospital, 28040 Madrid, Spain
| | - Javier Pozas
- Medical Oncology Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | | | | | | | - Jesús Chamorro-Pérez
- Medical Oncology Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
23
|
Efficacy of immune checkpoint inhibitor monotherapy or combined with other small molecule-targeted agents in ovarian cancer. Expert Rev Mol Med 2023; 25:e6. [PMID: 36691778 DOI: 10.1017/erm.2023.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ovarian cancer is the most lethal female reproductive system tumour. Despite the great advances in surgery and systemic chemotherapy over the past two decades, almost all patients in stages III and IV relapse and develop resistance to chemotherapy after first-line treatment. Ovarian cancer has an extraordinarily complex immunosuppressive tumour microenvironment in which immune checkpoints negatively regulate T cells activation and weaken antitumour immune responses by delivering immunosuppressive signals. Therefore, inhibition of immune checkpoints can break down the state of immunosuppression. Indeed, Immune checkpoint inhibitors (ICIs) have revolutionised the therapeutic landscape of many solid tumours. However, ICIs have yielded modest benefits in ovarian cancer. Therefore, a more comprehensive understanding of the mechanistic basis of the immune checkpoints is needed to improve the efficacy of ICIs in ovarian cancer. In this review, we systematically introduce the mechanisms and expression of immune checkpoints in ovarian cancer. Moreover, this review summarises recent updates regarding ICI monotherapy or combined with other small-molecule-targeted agents in ovarian cancer.
Collapse
|
24
|
Guo L, Kong D, Liu J, Zhan L, Luo L, Zheng W, Zheng Q, Chen C, Sun S. Breast cancer heterogeneity and its implication in personalized precision therapy. Exp Hematol Oncol 2023; 12:3. [PMID: 36624542 PMCID: PMC9830930 DOI: 10.1186/s40164-022-00363-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
Breast cancer heterogeneity determines cancer progression, treatment effects, and prognosis. However, the precise mechanism for this heterogeneity remains unknown owing to its complexity. Here, we summarize the origins of breast cancer heterogeneity and its influence on disease progression, recurrence, and therapeutic resistance. We review the possible mechanisms of heterogeneity and the research methods used to analyze it. We also highlight the importance of cell interactions for the origins of breast cancer heterogeneity, which can be further categorized into cooperative and competitive interactions. Finally, we provide new insights into precise individual treatments based on heterogeneity.
Collapse
Affiliation(s)
- Liantao Guo
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Deguang Kong
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Jianhua Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Ling Zhan
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Lan Luo
- Department of Breast Surgery, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Yunyan District, Guiyang, 550001, Guizhou, China
| | - Weijie Zheng
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| |
Collapse
|
25
|
Jarroudi OA, Bairi KE, Curigliano G, Afqir S. Immune-Checkpoint Inhibitors: A New Line of Attack in Triple-Negative Breast Cancer. Cancer Treat Res 2023; 188:29-62. [PMID: 38175341 DOI: 10.1007/978-3-031-33602-7_2] [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
Poor prognosis is a distinctive feature of triple-negative breast cancer (TNBC). Chemotherapy has long represented the main and unique treatment for patients with TNBC. Recently, immune checkpoint inhibitors (ICIs) were investigated in several clinical trials and were approved for clinical use in TNBC patients that express programmed cell death protein-1 (PD-1) in combination with chemotherapy in the first-line setting. ICIs are also being investigated in the neoadjuvant and adjuvant settings for TNBC. This chapter aims to discuss different ICIs used to treat all TNBC stages to date.
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
|
26
|
Huang X, Li XY, Shan WL, Chen Y, Zhu Q, Xia BR. Targeted therapy and immunotherapy: Diamonds in the rough in the treatment of epithelial ovarian cancer. Front Pharmacol 2023; 14:1131342. [PMID: 37033645 PMCID: PMC10080064 DOI: 10.3389/fphar.2023.1131342] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/21/2023] [Indexed: 04/11/2023] Open
Abstract
Currently, for ovarian cancer, which has the highest mortality rate among all gynecological cancers, the standard treatment protocol is initial tumor cytoreductive surgery followed by platinum-based combination chemotherapy. Although the survival rate after standard treatment has improved, the therapeutic effect of traditional chemotherapy is very limited due to problems such as resistance to platinum-based drugs and recurrence. With the advent of the precision medicine era, molecular targeted therapy has gradually entered clinicians' view, and individualized precision therapy has been realized, surpassing the limitations of traditional therapy. The detection of genetic mutations affecting treatment, especially breast cancer susceptibility gene (BRCA) mutations and mutations of other homologous recombination repair defect (HRD) genes, can guide the targeted drug treatment of patients, effectively improve the treatment effect and achieve a better patient prognosis. This article reviews different sites and pathways of targeted therapy, including angiogenesis, cell cycle and DNA repair, and immune and metabolic pathways, and the latest research progress from preclinical and clinical trials related to ovarian cancer therapy.
Collapse
Affiliation(s)
- Xu Huang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Bengbu Medical College Bengbu, Anhui, China
| | - Xiao-Yu Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Bengbu Medical College Bengbu, Anhui, China
| | - Wu-Lin Shan
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Qi Zhu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Bai-Rong Xia
- Bengbu Medical College Bengbu, Anhui, China
- *Correspondence: Bai-Rong Xia,
| |
Collapse
|
27
|
Skorda A, Bay ML, Hautaniemi S, Lahtinen A, Kallunki T. Kinase Inhibitors in the Treatment of Ovarian Cancer: Current State and Future Promises. Cancers (Basel) 2022; 14:6257. [PMID: 36551745 PMCID: PMC9777107 DOI: 10.3390/cancers14246257] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Ovarian cancer is the deadliest gynecological cancer, the high-grade serous ovarian carcinoma (HGSC) being its most common and most aggressive form. Despite the latest therapeutical advancements following the introduction of vascular endothelial growth factor receptor (VEGFR) targeting angiogenesis inhibitors and poly-ADP-ribose-polymerase (PARP) inhibitors to supplement the standard platinum- and taxane-based chemotherapy, the expected overall survival of HGSC patients has not improved significantly from the five-year rate of 42%. This calls for the development and testing of more efficient treatment options. Many oncogenic kinase-signaling pathways are dysregulated in HGSC. Since small-molecule kinase inhibitors have revolutionized the treatment of many solid cancers due to the generality of the increased activation of protein kinases in carcinomas, it is reasonable to evaluate their potential against HGSC. Here, we present the latest concluded and on-going clinical trials on kinase inhibitors in HGSC, as well as the recent work concerning ovarian cancer patient organoids and xenograft models. We discuss the potential of kinase inhibitors as personalized treatments, which would require comprehensive assessment of the biological mechanisms underlying tumor spread and chemoresistance in individual patients, and their connection to tumor genome and transcriptome to establish identifiable subgroups of patients who are most likely to benefit from a given therapy.
Collapse
Affiliation(s)
- Aikaterini Skorda
- Cancer Invasion and Resistance Group, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Marie Lund Bay
- Cancer Invasion and Resistance Group, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Sampsa Hautaniemi
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland
| | - Alexandra Lahtinen
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland
| | - Tuula Kallunki
- Cancer Invasion and Resistance Group, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
28
|
Nicolini A, Ferrari P, Carpi A. Immune Checkpoint Inhibitors and Other Immune Therapies in Breast Cancer: A New Paradigm for Prolonged Adjuvant Immunotherapy. Biomedicines 2022; 10:biomedicines10102511. [PMID: 36289773 PMCID: PMC9599105 DOI: 10.3390/biomedicines10102511] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/25/2022] [Accepted: 10/02/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Breast cancer is the most common form of cancer in women worldwide. Advances in the early diagnosis and treatment of cancer in the last decade have progressively decreased the cancer mortality rate, and in recent years, immunotherapy has emerged as a relevant tool against cancer. HER2+ and triple-negative breast cancers (TNBCs) are considered more immunogenic and suitable for this kind of treatment due to the higher rate of tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) expression. In TNBC, genetic aberrations further favor immunogenicity due to more neo-antigens in cancer cells. Methods: This review summarizes the principal ongoing conventional and investigational immunotherapies in breast cancer. Particularly, immune checkpoint inhibitors (ICIs) and their use alone or combined with DNA damage repair inhibitors (DDRis) are described. Then, the issue on immunotherapy with monoclonal antibodies against HER-2 family receptors is updated. Other investigational immunotherapies include a new schedule based on the interferon beta-interleukin-2 sequence that was given in ER+ metastatic breast cancer patients concomitant with anti-estrogen therapy, which surprisingly showed promising results. Results: Based on the scientific literature and our own findings, the current evaluation of tumor immunogenicity and the conventional model of adjuvant chemotherapy (CT) are questioned. Conclusions: A novel strategy based on additional prolonged adjuvant immunotherapy combined with hormone therapy or alternated with CT is proposed.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy
- Correspondence:
| | - Paola Ferrari
- Unit of Oncology, Department of Medical and Oncological Area, Azienda Ospedaliera-Universitaria Pisana, 56125 Pisa, Italy
| | - Angelo Carpi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
29
|
Xie T, Dickson KA, Yee C, Ma Y, Ford CE, Bowden NA, Marsh DJ. Targeting Homologous Recombination Deficiency in Ovarian Cancer with PARP Inhibitors: Synthetic Lethal Strategies That Impact Overall Survival. Cancers (Basel) 2022; 14:4621. [PMID: 36230543 PMCID: PMC9563432 DOI: 10.3390/cancers14194621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022] Open
Abstract
The advent of molecular targeted therapies has made a significant impact on survival of women with ovarian cancer who have defects in homologous recombination repair (HRR). High-grade serous ovarian cancer (HGSOC) is the most common histological subtype of ovarian cancer, with over 50% displaying defective HRR. Poly ADP ribose polymerases (PARPs) are a family of enzymes that catalyse the transfer of ADP-ribose to target proteins, functioning in fundamental cellular processes including transcription, chromatin remodelling and DNA repair. In cells with deficient HRR, PARP inhibitors (PARPis) cause synthetic lethality leading to cell death. Despite the major advances that PARPis have heralded for women with ovarian cancer, questions and challenges remain, including: can the benefits of PARPis be brought to a wider range of women with ovarian cancer; can other drugs in clinical use function in a similar way or with greater efficacy than currently clinically approved PARPis; what can we learn from long-term responders to PARPis; can PARPis sensitise ovarian cancer cells to immunotherapy; and can synthetic lethal strategies be employed more broadly to develop new therapies for women with ovarian cancer. We examine these, and other, questions with focus on improving outcomes for women with ovarian cancer.
Collapse
Affiliation(s)
- Tao Xie
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Kristie-Ann Dickson
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Christine Yee
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Yue Ma
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Caroline E. Ford
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Nikola A. Bowden
- Centre for Drug Repurposing and Medicines Research, University of Newcastle, Newcastle, NSW 2289, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2289, Australia
- Hunter Medical Research Institute, Newcastle, NSW 2289, Australia
| | - Deborah J. Marsh
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
30
|
PARP Inhibitors for Breast Cancer: Germline BRCA1/2 and Beyond. Cancers (Basel) 2022; 14:cancers14174332. [PMID: 36077867 PMCID: PMC9454726 DOI: 10.3390/cancers14174332] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/27/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Poly-adenosine diphosphate ribose polymerase (PARP) inhibitors (PARPi) are effective against tumors with mutations in DNA repair genes, most commonly in the BRCA1 and BRCA2 genes. Because these tumors are unable to repair their DNA, PARPi have been used to target DNA repair pathways and are useful in the treatment of breast cancers with some of these alterations. There are two FDA-approved PARPi for patients with breast cancer—olaparib and talazoparib. The data on olaparib and talazoparib in the treatment of breast cancer are summarized in this review, and we also explore potential future applications of PARPi beyond inherited BRCA mutations. Abstract Poly-adenosine diphosphate ribose polymerase (PARP) inhibitors (PARPi) are approved for BRCA1/2 carriers with HER2-negative breast cancer in the adjuvant setting with a high risk of recurrence as well as the metastatic setting. However, the indications for PARPi are broader for patients with other cancer types (e.g., prostate and ovarian cancer), involving additional biomarkers (e.g., ATM, PALB2, and CHEK) and genomic instability scores. Herein, we summarize the data on PARPi and breast cancer and discuss their use beyond BRCA carriers.
Collapse
|
31
|
Musacchio L, Cicala CM, Camarda F, Ghizzoni V, Giudice E, Carbone MV, Ricci C, Perri MT, Tronconi F, Gentile M, Salutari V, Scambia G, Lorusso D. Combining PARP inhibition and immune checkpoint blockade in ovarian cancer patients: a new perspective on the horizon? ESMO Open 2022; 7:100536. [PMID: 35849879 PMCID: PMC9294238 DOI: 10.1016/j.esmoop.2022.100536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/17/2022] [Accepted: 06/14/2022] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have completely reshaped the treatment of many malignancies, with remarkable improvements in survival outcomes. In ovarian cancer (OC), however, this emerging class of drugs has not yet found a favorable use due to results from phase I and II studies, which have not suggested a substantial antitumoral activity of these agents when administered as monotherapy. Robust preclinical data seem to suggest that the combination ICIs with poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) may result in a synergistic activity; furthermore, data from phase II clinical studies, evaluating this combination, have shown encouraging outcomes especially for those OC patients not suitable for platinum retreatment. While waiting for ongoing phase III clinical trial results, which will clarify the role of ICIs in combination with PARPis in the newly diagnosed OC, this review aims to summarize the preclinical data and clinical evidence available to date. Preclinical data indicate that PARPis exhibit immune modulating properties. The combination of PARPi with ICIs displays significant synergistic activity in preclinical models. Phase I and II clinical trials showed encouraging results for this combination, especially in platinum-resistant OC. Four ongoing phase III trials exploring the combination in first-line setting will delineate the role of immunotherapy in OC.
Collapse
Affiliation(s)
- L Musacchio
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy. https://twitter.com/lucia_musacchio
| | - C M Cicala
- Department of Medical and Surgical Science, Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy. https://twitter.com/carlomcicala
| | - F Camarda
- Department of Medical and Surgical Science, Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy. https://twitter.com/florianacamarda
| | - V Ghizzoni
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - E Giudice
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - M V Carbone
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - C Ricci
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - M T Perri
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - F Tronconi
- Medical Oncology Unit, Marche Polytechnic University, Ancona, Italy
| | - M Gentile
- Department of Biomedical Sciences and Human Oncology, University of Bari, Bari, Italy
| | - V Salutari
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - G Scambia
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Life Science and Public Health, Catholic University of Sacred Heart Largo Agostino Gemelli, Rome, Italy
| | - D Lorusso
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Life Science and Public Health, Catholic University of Sacred Heart Largo Agostino Gemelli, Rome, Italy.
| |
Collapse
|
32
|
McClurg DP, Urquhart G, McGoldrick T, Chatterji S, Miedzybrodzka Z, Speirs V, Elsberger B. Analysis of the Clinical Advancements for BRCA-Related Malignancies Highlights the Lack of Treatment Evidence for BRCA-Positive Male Breast Cancer. Cancers (Basel) 2022; 14:3175. [PMID: 35804947 PMCID: PMC9264767 DOI: 10.3390/cancers14133175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 12/10/2022] Open
Abstract
Male breast cancer (MBC) is a rare disease that accounts for less than 1% of all breast cancers and male malignancies. Despite recognised clinico-pathological and molecular differences to female breast cancer (FBC), the clinical management of MBC follows established FBC treatment strategies. Loss of function mutations in the DNA damage response genes BRCA1 and BRCA2, have been strongly implicated in the pathogenesis of MBC. While there have been extensive clinical advancements in other BRCA-related malignancies, including FBC, improvements in MBC remain stagnant. Here we present a review that highlights the lack of treatment evidence for BRCA-related MBC and the required national and global collaborative effort to address this unmet need. In doing so, we summarise the transformative clinical advancements with poly(ADP-ribose) polymerase (PARP) inhibitors in other BRCA-related cancers namely, FBC and prostate cancer.
Collapse
Affiliation(s)
- Dylan P. McClurg
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (D.P.M.); (S.C.); (Z.M.)
| | - Gordan Urquhart
- Aberdeen Royal Infirmary, Department of Oncology, Foresterhill Road, Aberdeen AB25 2ZN, UK; (G.U.); (T.M.)
| | - Trevor McGoldrick
- Aberdeen Royal Infirmary, Department of Oncology, Foresterhill Road, Aberdeen AB25 2ZN, UK; (G.U.); (T.M.)
| | - Subarnarekha Chatterji
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (D.P.M.); (S.C.); (Z.M.)
| | - Zosia Miedzybrodzka
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (D.P.M.); (S.C.); (Z.M.)
| | - Valerie Speirs
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (D.P.M.); (S.C.); (Z.M.)
| | - Beatrix Elsberger
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (D.P.M.); (S.C.); (Z.M.)
- Aberdeen Royal Infirmary, Breast Unit, Foresterhill Road, Aberdeen AB25 2ZN, UK
| |
Collapse
|
33
|
Gopinathan G, Berlato C, Lakhani A, Szabova L, Pegrum C, Pedrosa AR, Laforets F, Maniati E, Balkwill FR. Immune Mechanisms of Resistance to Cediranib in Ovarian Cancer. Mol Cancer Ther 2022; 21:1030-1043. [PMID: 35313341 PMCID: PMC9167758 DOI: 10.1158/1535-7163.mct-21-0689] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 11/16/2022]
Abstract
This article investigates mechanisms of resistance to the VEGF receptor inhibitor cediranib in high-grade serous ovarian cancer (HGSOC), and defines rational combination therapies. We used three different syngeneic orthotopic mouse HGSOC models that replicated the human tumor microenvironment (TME). After 4 to 5 weeks treatment of established tumors, cediranib had antitumor activity with increased tumor T-cell infiltrates and alterations in myeloid cells. However, continued cediranib treatment did not change overall survival or the immune microenvironment in two of the three models. Moreover, treated mice developed additional peritoneal metastases not seen in controls. Cediranib-resistant tumors had intrinsically high levels of IL6 and JAK/STAT signaling and treatment increased endothelial STAT3 activation. Combination of cediranib with a murine anti-IL6 antibody was superior to monotherapy, increasing mouse survival, reducing blood vessel density, and pSTAT3, with increased T-cell infiltrates in both models. In a third HGSOC model, that had lower inherent IL6 JAK/STAT3 signaling in the TME but high programmed cell death protein 1 (PD-1) signaling, long-term cediranib treatment significantly increased overall survival. When the mice eventually relapsed, pSTAT3 was still reduced in the tumors but there were high levels of immune cell PD-1 and Programmed death-ligand 1. Combining cediranib with an anti-PD-1 antibody was superior to monotherapy in this model, increasing T cells and decreasing blood vessel densities. Bioinformatics analysis of two human HGSOC transcriptional datasets revealed distinct clusters of tumors with IL6 and PD-1 pathway expression patterns that replicated the mouse tumors. Combination of anti-IL6 or anti-PD-1 in these patients may increase activity of VEGFR inhibitors and prolong disease-free survival.
Collapse
Affiliation(s)
- Ganga Gopinathan
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
| | - Chiara Berlato
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
| | - Anissa Lakhani
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
| | - Ludmila Szabova
- Frederick National Laboratory for Cancer Research, Tumour Microenvironment Leidos Biomedical Research Inc, Frederick, Maryland
| | - Colin Pegrum
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
| | - Ana-Rita Pedrosa
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
| | - Florian Laforets
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
| | - Eleni Maniati
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
| | - Frances R. Balkwill
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
| |
Collapse
|
34
|
Murphy AD, Morgan RD, Clamp AR, Jayson GC. The role of vascular endothelial growth factor inhibitors in the treatment of epithelial ovarian cancer. Br J Cancer 2022; 126:851-864. [PMID: 34716396 PMCID: PMC8927157 DOI: 10.1038/s41416-021-01605-5] [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: 05/10/2021] [Revised: 09/21/2021] [Accepted: 10/13/2021] [Indexed: 12/09/2022] Open
Abstract
Advanced epithelial ovarian, fallopian tube and primary peritoneal cancers (EOC) are a leading cause of gynaecological cancer-associated mortality and angiogenesis plays a key role in their growth. Vascular endothelial growth factor inhibitors (VEGFi) disrupt angiogenesis and improve the response rate, progression-free survival and in some cases, overall survival, when administered with and following cytotoxic chemotherapy, irrespective of the platinum sensitivity of EOC. Recent data have identified new indications for VEGFi in EOC: repeated exposure to VEGFi in the first- and then second-line treatment has sustained clinical efficacy; combinations of VEGFi with poly (ADP-ribose) polymerase inhibitors (PARPi) have proven effective as first-line or second-line maintenance regimens. However, recent trial data have not shown improved outcomes with combinations of VEGFi and immune checkpoint inhibitors. There remains a critical need to optimise patient selection for these effective yet somewhat toxic and expensive treatments. The search continues for validated biomarkers to optimise the use of VEGFi, of which the most promising at present is plasma Tie2. Based upon these studies, we propose a model of care incorporating VEGFi into the treatment of EOC, highlighting the need to change from the prescription of single courses of VEGFi, to allow use and re-use as clinically indicated.
Collapse
Affiliation(s)
| | - Robert D Morgan
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
- Division of Cancer Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Andrew R Clamp
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
- Division of Cancer Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Gordon C Jayson
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
- Division of Cancer Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| |
Collapse
|
35
|
Targeting DNA Damage Response and Immune Checkpoint for Anticancer Therapy. Int J Mol Sci 2022; 23:ijms23063238. [PMID: 35328658 PMCID: PMC8952261 DOI: 10.3390/ijms23063238] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023] Open
Abstract
Deficiency in DNA damage response (DDR) genes leads to impaired DNA repair functions that will induce genomic instability and facilitate cancer development. However, alterations of DDR genes can serve as biomarkers for the selection of suitable patients to receive specific therapeutics, such as immune checkpoint blockade (ICB) therapy. In addition, certain altered DDR genes can be ideal therapeutic targets through adapting the mechanism of synthetic lethality. Recent studies indicate that targeting DDR can improve cancer immunotherapy by modulating the immune response mediated by cGAS-STING-interferon signaling. Investigations of the interplay of DDR-targeting and ICB therapies provide more effective treatment options for cancer patients. This review introduces the mechanisms of DDR and discusses their crucial roles in cancer therapy based on the concepts of synthetic lethality and ICB. The contemporary clinical trials of DDR-targeting and ICB therapies in breast, colorectal, and pancreatic cancers are included.
Collapse
|
36
|
Giudice E, Gentile M, Salutari V, Ricci C, Musacchio L, Carbone MV, Ghizzoni V, Camarda F, Tronconi F, Nero C, Ciccarone F, Scambia G, Lorusso D. PARP Inhibitors Resistance: Mechanisms and Perspectives. Cancers (Basel) 2022; 14:1420. [PMID: 35326571 PMCID: PMC8945953 DOI: 10.3390/cancers14061420] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/27/2022] Open
Abstract
PolyADP-ribose polymerase (PARP) inhibitors (PARPis) represent the first clinically approved drugs able to provoke "synthetic lethality" in patients with homologous recombination-deficient (HRD) tumors. Four PARPis have just received approval for the treatment of several types of cancer. Besides, another three additional PARPis underlying the same mechanism of action are currently under investigation. Despite the success of these targeted agents, the increasing use of PARPis in clinical practice for the treatment of different tumors raised the issue of PARPis resistance, and the consequent disease relapse and dismal prognosis for patients. Several mechanisms of resistance have been investigated, and ongoing studies are currently focusing on strategies to address this challenge and overcome PARPis resistance. This review aims to analyze the mechanisms underlying PARPis resistance known today and discuss potential therapeutic strategies to overcome these processes of resistance in the future.
Collapse
Affiliation(s)
- Elena Giudice
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (E.G.); (V.G.); (G.S.)
| | - Marica Gentile
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Vanda Salutari
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (C.R.); (L.M.); (M.V.C.); (C.N.); (F.C.)
| | - Caterina Ricci
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (C.R.); (L.M.); (M.V.C.); (C.N.); (F.C.)
| | - Lucia Musacchio
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (C.R.); (L.M.); (M.V.C.); (C.N.); (F.C.)
| | - Maria Vittoria Carbone
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (C.R.); (L.M.); (M.V.C.); (C.N.); (F.C.)
| | - Viola Ghizzoni
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (E.G.); (V.G.); (G.S.)
| | - Floriana Camarda
- Medical Oncology, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy;
| | - Francesca Tronconi
- Medical Oncology, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy;
| | - Camilla Nero
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (C.R.); (L.M.); (M.V.C.); (C.N.); (F.C.)
| | - Francesca Ciccarone
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (C.R.); (L.M.); (M.V.C.); (C.N.); (F.C.)
| | - Giovanni Scambia
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (E.G.); (V.G.); (G.S.)
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (C.R.); (L.M.); (M.V.C.); (C.N.); (F.C.)
| | - Domenica Lorusso
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (E.G.); (V.G.); (G.S.)
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (C.R.); (L.M.); (M.V.C.); (C.N.); (F.C.)
| |
Collapse
|
37
|
Jiang G, Hong J, Shao F, Wen Q, Cheng F, Yu T, Zhu J. Evolution of Immunotherapy for Ovarian Cancer from a Bird's-Eye Perspective: A Text-Mining Analysis of Publication Trends and Topics. Front Oncol 2022; 12:795129. [PMID: 35280816 PMCID: PMC8907843 DOI: 10.3389/fonc.2022.795129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/24/2022] [Indexed: 11/24/2022] Open
Abstract
Objectives Ovarian tumors are among the most prominent gynecological malignancies and have a poor prognosis. Immunotherapy has undergone incredible progress in the past two decades. Our study aimed to use a bibliometric approach to identify research trends in ovarian cancer immunotherapy. Methods Literature on this topic published from 2000-2020 was retrieved from the Web of Science Core Citation database and analyzed using the bibliometric analysis software VOSviewer and CiteSpace. Results A total of 1729 articles on ovarian cancer immunotherapy published from January 2000 to December 2020 were identified. The number of published articles increased each year, from 40 in 2000 to 209 in 2020. These publications were from 61 countries, and the USA showed a dominant position in publication output, total citations, and average number of citations per paper. Co-citation networks revealed 14 subtopics. 'PD-L1 expression,' 'tumor reactive til,' and 'parp inhibitor' are the current potential subtopics. Furthermore, we determined research trends according to the timeline analysis. Conclusion Our study exhaustively describes the development and summarizes the research trends of ovarian cancer immunotherapy over the past 20 years.
Collapse
Affiliation(s)
- Guangyi Jiang
- Department of Gynecological Oncology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Junjie Hong
- Department of Gynecological Oncology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Feng Shao
- Department of Gynecological Oncology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Qiang Wen
- Department of Gynecological Oncology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Feng Cheng
- Department of Gynecological Oncology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Tunan Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianqing Zhu
- Department of Gynecological Oncology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| |
Collapse
|
38
|
Mutlu L, Harold J, Tymon-Rosario J, Santin AD. Immune checkpoint inhibitors for recurrent endometrial cancer. Expert Rev Anticancer Ther 2022; 22:249-258. [PMID: 35176955 DOI: 10.1080/14737140.2022.2044311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Endometrial cancer (EC) is the most common gynecologic malignancy. Outcomes for patients with advanced and/or recurrent disease have been modest with the use of chemotherapy. The approval of immune checkpoint inhibitors targeting PD-1 have recently revolutionized human cancer treatment. Recent trials with immune checkpoint inhibitors used alone or in combination with other agents, have demonstrated remarkable efficacy in the treatment of the all-comers EC patient population. AREAS COVERED In this article, we review major clinical trials on PD-1/PD-L1 inhibitors in advanced and recurrent EC and discuss the response rates to these agents in the context of their genomic background. EXPERT OPINION Immune checkpoint inhibitors have significantly changed our approach to the treatment of advanced/recurrent EC. Single agent anti-PD-1 regimens are highly effective in MMRd/MSI-H patients, but their clinical efficacy remains modest in MMR proficient/TMB low EC patients. Combination regimens that can decrease the tumor microenvironments immunosuppression and increase tumor immunogenicity represent a viable treatment option to broaden the activity of immune checkpoint inhibitors in advanced/recurrent EC patients. An increased understanding of the biomarkers of response and the molecular mechanisms of resistance to immune checkpoint inhibitors remains key for the next advancement of the field.
Collapse
Affiliation(s)
- Levent Mutlu
- 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
| | - Joan Tymon-Rosario
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| |
Collapse
|
39
|
Chilimoniuk Z, Rocka A, Stefaniak M, Tomczyk Ż, Jasielska F, Madras D, Filip A. Molecular methods for increasing the effectiveness of ovarian cancer treatment: a systematic review. Future Oncol 2022; 18:1627-1650. [PMID: 35129396 DOI: 10.2217/fon-2021-0565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: The aim of the current study is to analyze and summarize the latest research on improving therapy in ovarian cancer. Materials & methods: Data analysis was based on a review of publications from 2011 to 2021 in the PubMed database with use of the search terms including 'EGFR ovarian cancer', 'folate receptor inhibitors ovarian cancer', 'VEGF ovarian cancer', 'PDGF ovarian cancer' and 'CTLA-4 ovarian cancer'. Results: 6643 articles were found; 238 clinical trials and randomized control trials were analyzed; 122 studies were rejected due to inconsistency with the topic of the work. Conclusion: Extensive research on the treatment of ovarian cancer increases the chance of developing the most effective therapy suited to the individual needs of the patient.
Collapse
Affiliation(s)
- Zuzanna Chilimoniuk
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Agata Rocka
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Martyna Stefaniak
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Żaklina Tomczyk
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Faustyna Jasielska
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Dominika Madras
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Agata Filip
- Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| |
Collapse
|
40
|
Vafaei S, Zekiy AO, Khanamir RA, Zaman BA, Ghayourvahdat A, Azimizonuzi H, Zamani M. Combination therapy with immune checkpoint inhibitors (ICIs); a new frontier. Cancer Cell Int 2022; 22:2. [PMID: 34980128 PMCID: PMC8725311 DOI: 10.1186/s12935-021-02407-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022] Open
Abstract
Recently, immune checkpoint inhibitors (ICIs) therapy has become a promising therapeutic strategy with encouraging therapeutic outcomes due to their durable anti-tumor effects. Though, tumor inherent or acquired resistance to ICIs accompanied with treatment-related toxicities hamper their clinical utility. Overall, about 60-70% of patients (e.g., melanoma and lung cancer) who received ICIs show no objective response to intervention. The resistance to ICIs mainly caused by alterations in the tumor microenvironment (TME), which in turn, supports angiogenesis and also blocks immune cell antitumor activities, facilitating tumor cells' evasion from host immunosurveillance. Thereby, it has been supposed and also validated that combination therapy with ICIs and other therapeutic means, ranging from chemoradiotherapy to targeted therapies as well as cancer vaccines, can capably compromise tumor resistance to immune checkpoint blocked therapy. Herein, we have focused on the therapeutic benefits of ICIs as a groundbreaking approach in the context of tumor immunotherapy and also deliver an overview concerning the therapeutic influences of the addition of ICIs to other modalities to circumvent tumor resistance to ICIs.
Collapse
Affiliation(s)
- Somayeh Vafaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Angelina O. Zekiy
- Department of Prosthetic Dentistry, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ramadhan Ado Khanamir
- Internal Medicine and Surgery Department, College of Veterinary Medicine, University of Duhok, Kurdistan Region, Iraq
| | - Burhan Abdullah Zaman
- Basic Sciences Department, College of Pharmacy, University of Duhok, Kurdistan Region, Iraq
| | | | | | - Majid Zamani
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
41
|
Immuno-Oncology for Gynecologic Malignancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:193-232. [PMID: 34972966 DOI: 10.1007/978-3-030-79308-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Patients with advanced and/or recurrent gynecologic cancers derive limited benefit from currently available cytotoxic and targeted therapies. Successes of immunotherapy in other difficult-to-treat malignancies such as metastatic melanoma and advanced lung cancer have led to intense interest in clinical testing of these treatments in patients with gynecologic cancers. Currently, in the realm of gynecologic oncology, the FDA-approved use of immune checkpoint inhibitors is limited to microsatellite instability-high cancers, cancers with high tumor mutational burden, and PD-L1-positive cervical cancer. However, there has been an exponential growth of clinical trials testing immunotherapy approaches both alone and in combination with chemotherapy and/or targeted agents in patients with gynecologic cancers. This chapter will review some of the major reported and ongoing immunotherapy clinical trials in patients with endometrial, cervical, and epithelial ovarian cancer.
Collapse
|
42
|
Pham MM, Ngoi NYL, Peng G, Tan DSP, Yap TA. Development of poly(ADP-ribose) polymerase inhibitor and immunotherapy combinations: progress, pitfalls, and promises. Trends Cancer 2021; 7:958-970. [PMID: 34158277 PMCID: PMC8458234 DOI: 10.1016/j.trecan.2021.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/21/2022]
Abstract
The efficacy of poly(ADP-ribose) polymerase inhibitors (PARPi) is restricted by inevitable drug resistance, while their use in combination with chemotherapy and targeted agents is commonly associated with dose-limiting toxicities. Immune checkpoint blockade (ICB) has demonstrated durable responses in different solid tumors and is well-established across multiple cancers. Despite this, single agent activity is limited to a minority of patients and drug resistance remains an issue. Building on the monotherapy success of both drug classes, combining PARPi with ICB may be a safe and well-tolerated strategy with the potential to improve survival outcomes. In this review, we present the preclinical, translational, and clinical data supporting the combination of DNA damage response (DDR) and ICB as well as consider important questions to be addressed with future research.
Collapse
Affiliation(s)
- Melissa M Pham
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie Y L Ngoi
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Guang Peng
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S P Tan
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute, National University of Singapore, Singapore
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
43
|
Marchetti C, De Felice F, Romito A, Iacobelli V, Sassu CM, Corrado G, Ricci C, Scambia G, Fagotti A. Chemotherapy resistance in epithelial ovarian cancer: Mechanisms and emerging treatments. Semin Cancer Biol 2021; 77:144-166. [PMID: 34464704 DOI: 10.1016/j.semcancer.2021.08.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Ovarian cancer (OC) remains a fatal malignancy because most patients experience recurrent disease, which is resistant to chemotherapy. The outcomes for patients with platinum-resistant OC are poor, response rates to further chemotherapy are low and median survival is lower than 12 months. The complexity of platinum-resistant OC, which comprises a heterogeneous spectrum of diseases, is indeed far from being completely understood. Therefore, comprehending tumors' biological behaviour to identify reliable biomarkers, which may predict responses to therapies, is a demanding challenge to improve OC management. In the age of precision medicine, efforts to overcome platinum resistance in OC represent a dynamic and vast field in which innovative drugs and clinical trials rapidly develop. This review will present the exceptional biochemical environment implicated in OC and highlights mechanisms of chemoresistance. Furthermore, innovative molecules and new therapeutic opportunities are presented, along with currently available therapies and ongoing clinical trials.
Collapse
Affiliation(s)
- Claudia Marchetti
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.
| | - Francesca De Felice
- Division of Radiotherapy and Oncology, Policlinico Umberto I, Roma, Italy; Università La Sapienza, Roma, Italy
| | - Alessia Romito
- Gynecology and Breast Care Center, Mater Olbia Hospital, Olbia, Italy
| | - Valentina Iacobelli
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Department Woman and Child Health Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Carolina Maria Sassu
- Department of Maternal and Child Health and Urological Sciences, "Sapienza" University of Rome, Polyclinic Umberto I, Rome, Italy
| | - Giacomo Corrado
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Caterina Ricci
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Giovanni Scambia
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Department Woman and Child Health Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Anna Fagotti
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Department Woman and Child Health Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
44
|
Burguin A, Diorio C, Durocher F. Breast Cancer Treatments: Updates and New Challenges. J Pers Med 2021; 11:808. [PMID: 34442452 PMCID: PMC8399130 DOI: 10.3390/jpm11080808] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022] Open
Abstract
Breast cancer (BC) is the most frequent cancer diagnosed in women worldwide. This heterogeneous disease can be classified into four molecular subtypes (luminal A, luminal B, HER2 and triple-negative breast cancer (TNBC)) according to the expression of the estrogen receptor (ER) and the progesterone receptor (PR), and the overexpression of the human epidermal growth factor receptor 2 (HER2). Current BC treatments target these receptors (endocrine and anti-HER2 therapies) as a personalized treatment. Along with chemotherapy and radiotherapy, these therapies can have severe adverse effects and patients can develop resistance to these agents. Moreover, TNBC do not have standardized treatments. Hence, a deeper understanding of the development of new treatments that are more specific and effective in treating each BC subgroup is key. New approaches have recently emerged such as immunotherapy, conjugated antibodies, and targeting other metabolic pathways. This review summarizes current BC treatments and explores the new treatment strategies from a personalized therapy perspective and the resulting challenges.
Collapse
Affiliation(s)
- Anna Burguin
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada;
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
| | - Caroline Diorio
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
- Department of Preventive and Social Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada
| | - Francine Durocher
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada;
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
| |
Collapse
|
45
|
An D, Banerjee S, Lee JM. Recent advancements of antiangiogenic combination therapies in ovarian cancer. Cancer Treat Rev 2021; 98:102224. [PMID: 34051628 PMCID: PMC8217312 DOI: 10.1016/j.ctrv.2021.102224] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 12/19/2022]
Abstract
Ovarian cancer is a deadly malignancy with a growing therapeutic armamentarium, though achieving sustained benefit in the clinic remains largely elusive. Through biomarker and genetic analysis, several pathways of resistance and sensitivity to commonly used therapeutics have been identified, expanding the potential of identifying unique drug combinations and indicating new directions for improving clinical outcomes. Here, we review the mechanisms of angiogenic response and antiangiogenic therapy in ovarian cancer, as well as the interactions it exhibits with the immune and DNA damage response pathways. We discuss results from clinical trials examining the combinations of antiangiogenics, PARP inhibitors, and immune checkpoint inhibitors are also discussed, as well as several ongoing trials.
Collapse
Affiliation(s)
- Daniel An
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
46
|
Bäuerle T, Gupta S, Zheng S, Seyler L, Leporati A, Marosfoi M, Maschauer S, Prante O, Caravan P, Bogdanov A. Multimodal Bone Metastasis-associated Epidermal Growth Factor Receptor Imaging in an Orthotopic Rat Model. Radiol Imaging Cancer 2021; 3:e200069. [PMID: 34170199 DOI: 10.1148/rycan.2021200069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose To develop multimodality imaging techniques for measuring epidermal growth factor receptor (EGFR) as a therapy-relevant and metastasis-associated molecular marker in triple-negative mammary adenocarcinoma metastases. Materials and Methods An orthotopic bone metastasis EGFR-positive, triple-negative breast cancer (TNBC) model in rats was used for bioluminescence imaging, SPECT/CT, PET/CT, and MRI with quantitative analysis of transcripts (n = 22 rats). Receptor-specific MRI of EGFR expression in vivo was performed by acquiring spin-echo T1-weighted images after sequential administration of a pair of anti-EGFR antigen binding fragments, F(ab')2, conjugated to either horseradish peroxidase or glucose oxidase, which have complementing activities, as well as paramagnetic (gadolinium[III]-mono-5-hydroxytryptamide of 2,2',2''-(10-(2,6-dioxotetrahydro-2H-pyran-3-yl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl)triacetic acid, or Gd-5HT-DOTAGA) or positron-emitting (gallium 68-5HT-DOTAGA) substrates for MRI and PET/CT imaging, respectively. EGFR expression was confirmed by quantitative reverse transcriptase polymerase chain reaction and immunohistochemical analyses to compare with image findings. Results After surgical intraarterial delivery of TNBC cells, rats developed tumors that diverged into either rapidly growing osteolytic or slow-growing nonosteolytic tumors. Both tumor types showed receptor-specific initial MRI signal enhancement (contrast-to-noise ratio) that was three to six times higher than that of normal bone marrow (29.4 vs 4.9; P < .01). Micro PET/CT imaging of EGFR expression demonstrated a high level of heterogeneity with regional uptake of the tracer, which corresponded to region-of-interest MRI signal intensity elevation (121.1 vs 93.3; P < .001). Analysis of metastases with corroboration of imaging results showed high levels of EGFR protein and messenger RNA, or mRNA, expression in the invasive tumor. Conclusion Convergence of multimodal molecular receptor imaging enabled comprehensive assessment of EGFR overexpression in an orthotopic model of TNBC metastasis. Keywords: Animal Studies, Molecular Imaging-Cancer, MR-Contrast Agent, Radionuclide Studies, Skeletal-Appendicular, Metastases Supplemental material is available for this article. © RSNA, 2021.
Collapse
Affiliation(s)
- Tobias Bäuerle
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Suresh Gupta
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Shaokuan Zheng
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Lisa Seyler
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Anita Leporati
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Miklos Marosfoi
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Simone Maschauer
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Olaf Prante
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Peter Caravan
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| | - Alexei Bogdanov
- From the Institute of Radiology, Friedrich-Alexander University of Erlangen-Nurnberg, Erlangen, Germany (T.B., L.S.); Laboratory of Molecular Imaging Probes, Department of Radiology (S.G., A.L., A.B.), and Advanced MRI Center and New England Center for Stroke Research, Department of Radiology (S.Z., M.M.), University of Massachusetts Medical School, 55 Lake Ave North, S6-434, Worcester, MA 01655; Department of Nuclear Medicine, Friedrich-Alexander University of Erlangen-Nurnberg, Germany (S.M., O.P.); A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Mass (P.C.); and A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation (A.B.)
| |
Collapse
|
47
|
Shen K, Yang L, Li FY, Zhang F, Ding LL, Yang J, Lu J, Wang NN, Wang Y. Research progress of PARP inhibitor monotherapy and combination therapy for endometrial cancer. Curr Drug Targets 2021; 23:145-155. [PMID: 34139979 DOI: 10.2174/1389450122666210617111304] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/14/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022]
Abstract
Endometrial cancer is one of the three most common malignant tumors in the female reproductive system. Advanced and recurrent endometrial cancers have poor prognoses and lack effective treatments. Poly(ADP-ribose) polymerase (PARP) inhibitors have been applied to many different types of tumors, and they can selectively kill tumor cells that are defective in homologous recombination repair. Endometrial cancer is characterized by mutations in homologous recombination repair genes; accordingly, PARP inhibitors have achieved positive results in off-label treatments of endometrial cancer cases. Clinical trials of PARP inhibitors as monotherapies and within combination therapies for endometrial cancer are ongoing. For this review, we searched PubMed with "endometrial cancer" and "PARP inhibitor" as keywords, and we used "olaparib", "rucaparib", "niraparib" and "talazoparib" as search terms in clinicaltrials.gov for ongoing trials. The literature search ended in October 2020, and only English-language publications were selected. Multiple studies confirm that PARP inhibitors play an important role in killing tumor cells with defects in homologous recombination repair. Its combination with immune checkpoint inhibitors, PI3K/AKT/mTOR pathway inhibitors, cell cycle checkpoint inhibitors, and other drugs can improve the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Ke Shen
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| | - Li Yang
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| | - Fei-Yan Li
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| | - Feng Zhang
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| | - Lei-Lei Ding
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| | - Jing Yang
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| | - Jie Lu
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| | - Nan-Nan Wang
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| | - Yan Wang
- The Third Affiliated Hospital of Zhengzhou University, Obstetrics, and Gynecology, China
| |
Collapse
|
48
|
Ji X, Sui L, Song K, Lv T, Zhao H, Yao Q. PD-L1, PARP1, and MMRs as potential therapeutic biomarkers for neuroendocrine cervical cancer. Cancer Med 2021; 10:4743-4751. [PMID: 34076351 PMCID: PMC8290238 DOI: 10.1002/cam4.4034] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
Objective Neuroendocrine cervical cancer (NECC) is a rare cervical cancer with high aggressivity that causes poor prognosis even in the early stage. Given other neuroendocrine carcinomas and other types of cervical cancer have been proved to have expression of programmed cell death protein 1 ligand 1(PD‐L1) and poly ADP‐ribose polymerase‐1(PARP1), we would measure and analyze these proteins in this invasive cancer. The purpose of this study is to investigate the application value of PD‐1/PD‐L1 and PARP1 inhibitors in NECC. Methods The NECC cases in our center with formalin‐fixed paraffin‐embedded tissue blocks were collected, and immunohistochemical (IHC) staining of PD‐L1, PARP1, Mismatch repair proteins (MMRs), and P53 was performed. Chi‐square test was used to analyze associations between various protein expressions. We analyzed the efficacy of immunotherapy in a recent patient with secondary recurrence after two courses of chemotherapy. Results After rigorous screening, 20 cases were finally included. Three cases did not undergo surgical treatment because of their advanced stage. Twelve (60%) developed distant metastases or relapsed within five years, and most of them within two years. The positive rate of PD‐L1 and PARP1 were 70% and 75% respectively. Among all the cases, microsatellite instability (MSI) was seen in six cases (30%) and abnormal p53 expression was in 15 patients (75%). PD‐L1 was associated with PARP1 expression in the MSI subgroup. The patient treated with chemotherapy + VEGF inhibitor (VEGFi) + programmed cell death protein 1(PD‐1) inhibitor had an excellent improvement in clinical symptoms, tumor markers, and mass size. Conclusion The IHC results of PD‐L1, PARP1, and MMRs suggested that NECC was the target of immunotargeted therapy. Our case confirmed that immune checkpoint therapy was effective in patients with PD‐L1 positive and MMRs loss. Considering the clinical practicability, more cases should be collected, and effective biomarkers still need to be further searched.
Collapse
Affiliation(s)
- Xiaoyu Ji
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Sui
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kejuan Song
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Teng Lv
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Han Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qin Yao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Varayathu H, Sarathy V, Thomas BE, Mufti SS, Naik R. Combination Strategies to Augment Immune Check Point Inhibitors Efficacy - Implications for Translational Research. Front Oncol 2021; 11:559161. [PMID: 34123767 PMCID: PMC8193928 DOI: 10.3389/fonc.2021.559161] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 04/30/2021] [Indexed: 12/22/2022] Open
Abstract
Immune checkpoint inhibitor therapy has revolutionized the field of cancer immunotherapy. Even though it has shown a durable response in some solid tumors, several patients do not respond to these agents, irrespective of predictive biomarker (PD-L1, MSI, TMB) status. Multiple preclinical, as well as early-phase clinical studies are ongoing for combining immune checkpoint inhibitors with anti-cancer and/or non-anti-cancer drugs for beneficial therapeutic interactions. In this review, we discuss the mechanistic basis behind the combination of immune checkpoint inhibitors with other drugs currently being studied in early phase clinical studies including conventional chemotherapy drugs, metronomic chemotherapy, thalidomide and its derivatives, epigenetic therapy, targeted therapy, inhibitors of DNA damage repair, other small molecule inhibitors, anti-tumor antibodies hormonal therapy, multiple checkpoint Inhibitors, microbiome therapeutics, oncolytic viruses, radiotherapy, drugs targeting myeloid-derived suppressor cells, drugs targeting Tregs, drugs targeting renin-angiotensin system, drugs targeting the autonomic nervous system, metformin, etc. We also highlight how translational research strategies can help better understand the true therapeutic potential of such combinations.
Collapse
Affiliation(s)
- Hrishi Varayathu
- Department of Translational Medicine and Therapeutics, HealthCare Global Enterprises Limited, Bangalore, India
| | - Vinu Sarathy
- Department of Medical Oncology, HealthCare Global Enterprises Limited, Bangalore, India
| | - Beulah Elsa Thomas
- Department of Clinical Pharmacology, HealthCare Global Enterprises Limited, Bangalore, India
| | - Suhail Sayeed Mufti
- Department of Translational Medicine and Therapeutics, HealthCare Global Enterprises Limited, Bangalore, India
| | - Radheshyam Naik
- Department of Medical Oncology, HealthCare Global Enterprises Limited, Bangalore, India
| |
Collapse
|
50
|
Majidpoor J, Mortezaee K. The efficacy of PD-1/PD-L1 blockade in cold cancers and future perspectives. Clin Immunol 2021; 226:108707. [DOI: 10.1016/j.clim.2021.108707] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/15/2021] [Accepted: 02/25/2021] [Indexed: 12/30/2022]
|