1
|
Raman R, Debata S, Govindarajan T, Kumar P. Targeting Triple-Negative Breast Cancer: Resistance Mechanisms and Therapeutic Advancements. Cancer Med 2025; 14:e70803. [PMID: 40318146 PMCID: PMC12048392 DOI: 10.1002/cam4.70803] [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: 07/10/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is one of the most heterogeneous and menacing forms of breast cancer, with no sustainable cure available in the current treatment landscape. Its lack of targets makes it highly unresponsive to various treatment modalities, which is why chemotherapy continues to be the primary form of treatment, despite the high rates of patients developing chemoresistance. In recent years, however, there has been significant progress in identifying and understanding the role of several aspects that might contribute to genomic instability and other hallmarks of cancer, including cellular proteins, immune targets, and epigenetic mechanisms, which are desirable as they permit reversibility easier than the often-adamant genetic changes. METHODS A literature review was conducted on the role of various TNBC associated biomarkers, their therapeutic applications, and their role in tumorigenesis and tumor maintenance, with a focus on linking both the driving biological mechanisms and emerging treatment options for TNBC. CONCLUSIONS Shifting the focus of treatment to identify crucial tumor cell subpopulations and associated biomarkers, such as local immune cell populations and cancer stem cells, could potentially solve or simplify decades' worth of problems that are associated with TNBC, bolstering early detection and the evolution of precision medicine and treatment. The techniques that can be used here are epigenetic analysis and RNA sequencing. Biomarkers, such as PD-L1, survivin, and ABC transporters, are implicated in several crucial processes that maintain tumors, such as cell proliferation, metastasis, immunosuppression, and stemness. Complex treatment options such as, immunotherapy, pathway inhibition, PARP inhibition, virotherapy, and RNA targeting have been considered for TNBC. Phytochemicals are also being considered as a treatment modality for TNBC, as a supplement to chemotherapy and radiation therapy, or as sole treatment.
Collapse
Affiliation(s)
- Rachana Raman
- Photoceutics and Regeneration Laboratory, Department of Biotechnology, Centre for Microfluidics, Biomarkers, Photoceutics and Sensors (μBioPS), Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
- Innotech Manipal, Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
| | - Shristi Debata
- Department of Biotechnology, Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
| | | | - Praveen Kumar
- Photoceutics and Regeneration Laboratory, Department of Biotechnology, Centre for Microfluidics, Biomarkers, Photoceutics and Sensors (μBioPS), Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
- Innotech Manipal, Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
| |
Collapse
|
2
|
Sun P, Ding Z, Chen J, Ou K, Zhou D, Li R, Gu T, Sun H, Cheng Y. Prognostic characteristics and drug sensitivity analysis of hepatocellular carcinoma based on histone modification-related genes: a multi-omics integrated study revealing potential therapeutic targets and individualized treatment strategies. Front Pharmacol 2024; 15:1489469. [PMID: 39584133 PMCID: PMC11582355 DOI: 10.3389/fphar.2024.1489469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) ranks among the most prevalent and lethal malignancies worldwide. Histone modifications (HMs) play a pivotal role in the initiation and progression of HCC. However, our understanding of HMs in HCC remains limited due to the disease's heterogeneity and the complexity of HMs. Methods We integrated multi-omics data from multiple cohorts, including single-cell RNA sequencing, bulk RNA sequencing, and clinical information. Weighted gene co-expression network analysis (WGCNA) and consensus clustering were employed to identify histone-related genes. We developed a histone modification-related signature (HMRS) using 117 machine learning methods. Comprehensive analyses of molecular characteristics, immune landscape, and drug sensitivity associated with the HMRS were performed. Results Through integrative analysis, we defined 110 histone-related genes and identified 45 HCC-HM-related genes (HCC-HMRgenes). The HMRS demonstrated robust prognostic value across multiple cohorts. Patients with high HMRS scores exhibited distinct genomic alterations, including higher tumor heterogeneity and TP53 mutations. The high-risk group showed enrichment in cell cycle, DNA repair, and metabolic pathways. Immune landscape analysis revealed significant differences in immune cell infiltration and pathway activities between high- and low-risk groups. Drug sensitivity prediction suggested potential therapeutic strategies for different risk groups. Conclusion Our study provides a comprehensive understanding of HMs in HCC and establishes a robust prognostic signature. The HMRS not only stratifies patients into distinct risk groups but also offers insights into underlying molecular mechanisms, immune characteristics, and potential therapeutic strategies, paving the way for personalized medicine in HCC.
Collapse
Affiliation(s)
- Ping Sun
- Department of Organ transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Lab Organ Transplantation of Liaoning Province, Shenyang, Liaoning, China
| | - Zheng Ding
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Juan Chen
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Kezhen Ou
- Department of Organ transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Lab Organ Transplantation of Liaoning Province, Shenyang, Liaoning, China
| | - Dianjie Zhou
- Department of Organ transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Lab Organ Transplantation of Liaoning Province, Shenyang, Liaoning, China
| | - Rui Li
- Department of Organ transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Lab Organ Transplantation of Liaoning Province, Shenyang, Liaoning, China
| | - Tianxiang Gu
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Sun
- Department of Organ transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Lab Organ Transplantation of Liaoning Province, Shenyang, Liaoning, China
| | - Ying Cheng
- Department of Organ transplantation and Hepatobiliary, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Lab Organ Transplantation of Liaoning Province, Shenyang, Liaoning, China
| |
Collapse
|
3
|
Cheng F, Zhang Y, Xiong H, Zhao M, Wang Q, Zhu Y, Li Y, Tang R, Li J. NMNATs expression inhibition mediated NAD + deficiency plays a critical role in doxorubicin-induced hepatotoxicity in mice. Toxicol Appl Pharmacol 2024; 482:116799. [PMID: 38160893 DOI: 10.1016/j.taap.2023.116799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/06/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Doxorubicin (DOX) is one of the most widely used antineoplastic drugs with known cardiotoxicity while other organ toxicity, such as hepatotoxicity is not well defined. This study was to explore the role of nicotinamide adenine dinucleotide (NAD+) in DOX-induced hepatotoxicity. DOX (20 mg/kg) induced acute liver injury and oxidative stress in C57BL/6 J mice at 48 h. Notably, the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and NAD(P)H dehydrogenase quinone 1 (NQO1) were downregulated. NAD+ deficiency was confirmed due to DOX exposure. Mechanistically, the downregulation of nicotinamide mononucleotide adenylyl transferase 1 (NMNAT1), NMNAT2 and NMNAT3, while no alteration of nicotinamide phosphoribosyl transferase was proved. As a consequence of NAD+ deficiency, the expression of poly-ADP-ribose polymerase1 (PARP1), CD38 and Sirtuin1 (SIRT1) were reduced. Furthermore, supplementation of NAD+ (200 mg/kg/day) or its precursor nicotinamide mononucleotide (NMN) (500 mg/kg/day) alleviated liver injury, attenuated oxidative stress, and elevated the downregulation of Nrf2 and NQO1. More importantly, compromised expression of NMNAT1-3, PARP1, CD38 and SIRT1 were improved by NAD+ and NMN. In conclusion, NAD+ deficiency due to NMNATs expression inhibition may attribute to the pathogenesis of DOX-induced hepatotoxicity, thus providing new insights for mitigating DOX side effects.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China
| | - Yongtai Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China
| | - Hongli Xiong
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China
| | - Minzhu Zhao
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China
| | - Qi Wang
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China
| | - Ying Zhu
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China
| | - Yongguo Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China
| | - Renkuan Tang
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China
| | - Jianbo Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, PR China; Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing, PR China; Chongqing Key Laboratory of Forensic Medicine, Chongqing, PR China.
| |
Collapse
|
4
|
Yu S, Xiao H, Ma L, Zhang J, Zhang J. Reinforcing the immunogenic cell death to enhance cancer immunotherapy efficacy. Biochim Biophys Acta Rev Cancer 2023; 1878:188946. [PMID: 37385565 DOI: 10.1016/j.bbcan.2023.188946] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Immunogenic cell death (ICD) has been a revolutionary modality in cancer treatment since it kills primary tumors and prevents recurrent malignancy simultaneously. ICD represents a particular form of cancer cell death accompanied by production of damage-associated molecular patterns (DAMPs) that can be recognized by pattern recognition receptors (PRRs), which enhances infiltration of effector T cells and potentiates antitumor immune responses. Various treatment methods can elicit ICD involving chemo- and radio-therapy, phototherapy and nanotechnology to efficiently convert dead cancer cells into vaccines and trigger the antigen-specific immune responses. Nevertheless, the efficacy of ICD-induced therapies is restrained due to low accumulation in the tumor sites and damage of normal tissues. Thus, researchers have been devoted to overcoming these problems with novel materials and strategies. In this review, current knowledge on different ICD modalities, various ICD inducers, development and application of novel ICD-inducing strategies are summarized. Moreover, the prospects and challenges are briefly outlined to provide reference for future design of novel immunotherapy based on ICD effect.
Collapse
Affiliation(s)
- Sihui Yu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Hongyang Xiao
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Li Ma
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Jiarong Zhang
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
Xu H, Huang K, Lin Y, Gong H, Ma X, Zhang D. Glycosyltransferase GLT8D1 and GLT8D2 serve as potential prognostic biomarkers correlated with Tumor Immunity in Gastric Cancer. BMC Med Genomics 2023; 16:123. [PMID: 37277853 PMCID: PMC10242987 DOI: 10.1186/s12920-023-01559-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/27/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Glycosylation involved in various biological function, aberrant glycosylation plays an important role in cancer development and progression. Glycosyltransferase 8 domain containing 1 (GLT8D1) and GLT8D2, as members of the glycosyltransferase family proteins, are associated with transferase activity. However, the association between GLT8D1/2 and gastric cancer (GC) remains unclear. We aimed to investigate the potential prognostic value and oncogenic role of GLT8D1/2 in GC. METHODS The relationship between GLT8D1/2 and GC was evaluated through comprehensive bioinformatics approaches. A series of factors like gene expression patterns, Kaplan-Meier survival analyses, Cox regression analyses, prognostic nomogram, calibration curves, ROC curves, function enrichment analyses, tumor immunity association, genetic alterations, and DNA methylation were included. Data and statistical analyses were performed using R software (v3.6.3). RESULTS Both GLT8D1 and GLT8D2 expression were significantly upregulated in GC tissues(n = 414) compared with normal tissues(n = 210), and high expression of GLT8D1/2 was remarkably correlated with poor prognosis for GC patients. Cox regression analyses implied that GLT8D1/2 could act as independent prognostic factors in GC. Furthermore, gene function analyses indicated that multiple signaling pathways involving tumor oncogenesis and development enriched, such as mTOR, cell cycle, MAPK, Notch, Hedgehog, FGF, and PI3K-Akt signaling pathways. Moreover, GLT8D1/2 was significantly associated with immune cell infiltration, immune checkpoint genes, and immune regulators TMB/MSI. CONCLUSION GLT8D1/2 may serve as potential prognostic markers of poor prognosis in GC correlated with tumor immunity. The study provided an insight into identifying potential biomarkers and targets for prognosis, immunotherapy response, and therapy in GC.
Collapse
Affiliation(s)
- Huimei Xu
- Department of Gastroenterology, The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, P.R. China
- Lanzhou University Second Hospital, Lanzhou, 730030, P.R. China
| | - Ke Huang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730030, P.R. China
| | - Yimin Lin
- Department of Gastroenterology, The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, P.R. China
- Lanzhou University Second Hospital, Lanzhou, 730030, P.R. China
| | - Hang Gong
- Department of Gastroenterology, The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, P.R. China
- Lanzhou University Second Hospital, Lanzhou, 730030, P.R. China
| | - Xueni Ma
- Department of Gastroenterology, The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, P.R. China
- Lanzhou University Second Hospital, Lanzhou, 730030, P.R. China
| | - Dekui Zhang
- Lanzhou University Second Hospital, Lanzhou, 730030, P.R. China.
- Key Laboratory of Digestive Diseases of Lanzhou University Second Hospital, Lanzhou, 730030, P.R. China.
| |
Collapse
|
6
|
Paul S, Sinha S, Kundu CN. Targeting cancer stem cells in the tumor microenvironment: An emerging role of PARP inhibitors. Pharmacol Res 2022; 184:106425. [PMID: 36075511 DOI: 10.1016/j.phrs.2022.106425] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/19/2022]
Abstract
Cancer stem cells (CSCs) constitute a small population of cancer cells in the tumor microenvironment (TME), which are responsible for metastasis, angiogenesis, drug resistance, and cancer relapse. Understanding the key signatures and resistance mechanisms of CSCs may help in the development of novel chemotherapeutic strategies to specifically target CSCs in the TME. PARP inhibitors (PARPi) are known to enhance the chemosensitivity of cancer cells to other chemotherapeutic agents by inhibiting the DNA repair pathways and chromatin modulation. But their effects on CSCs are still unknown. Few studies have reported that PARPi can stall replication fork progression in CSCs. PARPi also have the potential to overcome chemoresistance in CSCs and anti-angiogenic potentiality as well. Previous reports have suggested that epigenetic drugs can synergistically ameliorate the anti-cancer activities of PARPi through epigenetic modulations. In this review, we have systematically discussed the effects of PARPi on different DNA repair pathways with respect to CSCs and also how CSCs can be targeted either as monotherapy or as a part of combination therapy. We have also talked about how PARPi can help in reversal of chemoresistance of CSCs and the role of PARPi in epigenetic modifications to hinder cancer progression. We have also elaborated on the aspects of research that need to be investigated for development of successful therapeutic interventions using PARPi to specifically target CSCs in the TME.
Collapse
Affiliation(s)
- Subarno Paul
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Saptarshi Sinha
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
7
|
Prasanna T, Malik L, McCuaig RD, Tu WJ, Wu F, Lim PS, Tan AHY, Dahlstrom JE, Clingan P, Moylan E, Chrisp J, Fuller D, Rao S, Yip D. A Phase 1 Proof of Concept Study Evaluating the Addition of an LSD1 Inhibitor to Nab-Paclitaxel in Advanced or Metastatic Breast Cancer (EPI-PRIMED). Front Oncol 2022; 12:862427. [PMID: 35719960 PMCID: PMC9205212 DOI: 10.3389/fonc.2022.862427] [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: 01/25/2022] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Lysine-Specific Demethylase-1 (LSD1) is overexpressed in breast cancer cells and facilitate mesenchymal properties which may contribute to therapeutic resistance and cancer progression. The purpose of this study was to investigate the safety of combination, nab-paclitaxel and phenelzine, an irreversible LSD1 inhibitor in patients with metastatic breast cancer (mBC). METHODS Eligible patients with mBC were treated with nab-paclitaxel (100mg/m2) weekly for 3 weeks with one week break in a 28-day cycle. Dose escalation of phenelzine followed the Cumulative Cohort Design and phenelzine treatment commenced from day 2 of first cycle. Eleven patients were screened, and eligible patients were enrolled in cohorts with the dose of phenelzine ranging from 45mg to 90mg. RESULTS The Optimum Biological Dose was established at 60mg of phenelzine daily in combination with nab-paclitaxel and considered as the recommended phase 2 dose. Most (95%) of adverse events were grade 1 or 2 with two grade 3 events being diarrhea and neutropenia at 45mg and 60mg phenelzine respectively, with no unexpected toxicity/deaths. Commonly reported toxicities were fatigue (n=4,50%), dizziness (n=6,75%), neutropenia (n=3,37.5%), peripheral neuropathy (n=3,37.5%), diarrhea (n=2,25%), and hallucination (n=2,25%). After a median follow up of 113 weeks, all patients showed disease progression on trial with 4 patients being alive at the time of data cut off, including one patient with triple negative breast cancer. Median progression-free survival was 34 weeks. Significant inhibition of LSD1 and suppression of mesenchymal markers in circulating tumor cells were noted. CONCLUSION Phenelzine in combination with nab-paclitaxel was well tolerated, without any unexpected toxicities in patients with mBC and demonstrated evidence of antitumor activity. For the first time, this proof-of-concept study showed in-vivo inhibition of LSD1 suppressed mesenchymal markers, which are known to facilitate generation of cancer stem cells with metastatic potential. Clinical Trial Registration: ClinicalTrials.Gov NCT03505528, UTN of U1111-1197-5518.
Collapse
Affiliation(s)
- Thiru Prasanna
- Australian National University (ANU) Medical School, Australian National University, Canberra, ACT, Australia
- Department of Medical Oncology, The Canberra Hospital, Garran, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Bruce, ACT, Australia
| | - Laeeq Malik
- Australian National University (ANU) Medical School, Australian National University, Canberra, ACT, Australia
- Department of Medical Oncology, The Canberra Hospital, Garran, ACT, Australia
| | - Robert D. McCuaig
- Gene Regulation and Translational Medicine Laboratory, Infection and Inflammation Program, Queensland Institute of Medical Research Berghofer, Brisbane, QLD, Australia
| | - Wen Juan Tu
- Gene Regulation and Translational Medicine Laboratory, Infection and Inflammation Program, Queensland Institute of Medical Research Berghofer, Brisbane, QLD, Australia
| | - Fan Wu
- Faculty of Science and Technology, University of Canberra, Bruce, ACT, Australia
| | - Pek Siew Lim
- Faculty of Science and Technology, University of Canberra, Bruce, ACT, Australia
| | - Abel H. Y. Tan
- Faculty of Science and Technology, University of Canberra, Bruce, ACT, Australia
| | - Jane E. Dahlstrom
- Australian National University (ANU) Medical School, Australian National University, Canberra, ACT, Australia
- Department of Anatomical Pathology, ACT Pathology, The Canberra Hospital, Canberra, ACT, Australia
| | - Philip Clingan
- Department of Medical Oncology, Southern Medical Day Care Centre, Wollongong, NSW, Australia
| | - Eugene Moylan
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW, Australia
| | - Jeremy Chrisp
- EpiAxis Therapeutics Pty Ltd, Sydney, NSW, Australia
| | - David Fuller
- EpiAxis Therapeutics Pty Ltd, Sydney, NSW, Australia
| | - Sudha Rao
- Gene Regulation and Translational Medicine Laboratory, Infection and Inflammation Program, Queensland Institute of Medical Research Berghofer, Brisbane, QLD, Australia
| | - Desmond Yip
- Australian National University (ANU) Medical School, Australian National University, Canberra, ACT, Australia
- Department of Medical Oncology, The Canberra Hospital, Garran, ACT, Australia
| |
Collapse
|
8
|
Zhao L, Guo H, Chen X, Zhang W, He Q, Ding L, Yang B. Tackling drug resistance in ovarian cancer with epigenetic targeted drugs. Eur J Pharmacol 2022; 927:175071. [PMID: 35636522 DOI: 10.1016/j.ejphar.2022.175071] [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: 11/30/2021] [Revised: 05/11/2022] [Accepted: 05/24/2022] [Indexed: 12/26/2022]
Abstract
Epigenetic dysregulation plays a crucial role in the development and progression of ovarian cancer. Since the first experiment conducted on resistant ovarian cancer cells using demethylating drugs, multiple clinical trials have revealed that epigenetic targeted drugs combined with chemotherapy, molecular-targeted drugs, or even immunotherapy could enhance tumor sensitivity and reverse acquired resistances. Here, we summarized the combination strategies of epigenetic targeted drugs with other treatment strategies of ovarian cancer and discussed the principles of combination therapy. Finally, we enumerated several reasonable clinical trial designs as well as future drug development strategies, which may provide promising ideas for the application of epigenetic drugs to ovarian cancer.
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
| | - Hongjie Guo
- 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
| | - Wenxin Zhang
- 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, China; Cancer Center of Zhejiang University, Hangzhou, 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, China; Cancer Center of Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Zhu L, Liu J, Chen J, Zhou Q. The developing landscape of combinatorial therapies of immune checkpoint blockade with DNA damage repair inhibitors for the treatment of breast and ovarian cancers. J Hematol Oncol 2021; 14:206. [PMID: 34930377 PMCID: PMC8686226 DOI: 10.1186/s13045-021-01218-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023] Open
Abstract
The use of immune checkpoint blockade (ICB) using antibodies against programmed death receptor (PD)-1, PD ligand (PD-L)-1, and cytotoxic T-lymphocyte antigen 4 (CTLA-4) has redefined the therapeutic landscape in solid tumors, including skin, lung, bladder, liver, renal, and breast tumors. However, overall response rates to ICB therapy remain limited in PD-L1-negative patients. Thus, rational and effective combination therapies will be needed to address ICB treatment resistance in these patients, as well as in PD-L1-positive patients who have progressed under ICB treatment. DNA damage repair inhibitors (DDRis) may activate T-cell responses and trigger inflammatory cytokines release and eventually immunogenic cancer cell death by amplifying DNA damage and generating immunogenic neoantigens, especially in DDR-defective tumors. DDRi may also lead to adaptive PD-L1 upregulation, providing a rationale for PD-L1/PD-1 blockade. Thus, based on preclinical evidence of efficacy and no significant overlapping toxicity, some ICB/DDRi combinations have rapidly progressed to clinical testing in breast and ovarian cancers. Here, we summarize the available clinical data on the combination of ICB with DDRi agents for treating breast and ovarian cancers and discuss the mechanisms of action and other lessons learned from translational studies conducted to date. We also review potential biomarkers to select patients most likely to respond to ICB/DDRi combination therapy.
Collapse
Affiliation(s)
- Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jiewei Liu
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jiang Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| | - Qinghua Zhou
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
10
|
Wang M, Chen S, Ao D. Targeting DNA repair pathway in cancer: Mechanisms and clinical application. MedComm (Beijing) 2021; 2:654-691. [PMID: 34977872 PMCID: PMC8706759 DOI: 10.1002/mco2.103] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 02/05/2023] Open
Abstract
Over the last decades, the growing understanding on DNA damage response (DDR) pathways has broadened the therapeutic landscape in oncology. It is becoming increasingly clear that the genomic instability of cells resulted from deficient DNA damage response contributes to the occurrence of cancer. One the other hand, these defects could also be exploited as a therapeutic opportunity, which is preferentially more deleterious in tumor cells than in normal cells. An expanding repertoire of DDR-targeting agents has rapidly expanded to inhibitors of multiple members involved in DDR pathways, including PARP, ATM, ATR, CHK1, WEE1, and DNA-PK. In this review, we sought to summarize the complex network of DNA repair machinery in cancer cells and discuss the underlying mechanism for the application of DDR inhibitors in cancer. With the past preclinical evidence and ongoing clinical trials, we also provide an overview of the history and current landscape of DDR inhibitors in cancer treatment, with special focus on the combination of DDR-targeted therapies with other cancer treatment strategies.
Collapse
Affiliation(s)
- Manni Wang
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Siyuan Chen
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Danyi Ao
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
11
|
Chiorean EG, Guthrie KA, Philip PA, Swisher EM, Jalikis F, Pishvaian MJ, Berlin J, Noel MS, Suga JM, Garrido-Laguna I, Cardin DB, Radke MR, Duong M, Bellasea S, Lowy AM, Hochster HS. Randomized Phase II Study of PARP Inhibitor ABT-888 (Veliparib) with Modified FOLFIRI versus FOLFIRI as Second-line Treatment of Metastatic Pancreatic Cancer: SWOG S1513. Clin Cancer Res 2021; 27:6314-6322. [PMID: 34580114 PMCID: PMC8639715 DOI: 10.1158/1078-0432.ccr-21-1789] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/08/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE PARP inhibitors synergize with topoisomerase inhibitors, and veliparib plus modified (m) FOLFIRI (no 5-FU bolus) had preliminary activity in metastatic pancreatic cancers. This study evaluated the safety and efficacy of second-line treatment with veliparib and mFOLFIRI versus FOLFIRI (control) for metastatic pancreatic cancer. PATIENTS AND METHODS This randomized phase II clinical trial led by the SWOG Cancer Research Network enrolled patients between September 1, 2016 and December 13, 2017. The median follow-up was 9 months (IQR 1-27). BRCA1/2 and homologous recombination DNA damage repair (HR-DDR) genetic defects were tested in blood and tumor biopsies. Patients received veliparib 200 mg twice daily, days 1-7 with mFOLFIRI days 3-5, or FOLFIRI in 14-day cycles. RESULTS After 123 of planned 143 patients were accrued, an interim futility analysis indicated that the veliparib arm was unlikely to be superior to control, and the study was halted. Median overall survival (OS) was 5.4 versus 6.5 months (HR, 1.23; P = 0.28), and median progression-free survival (PFS) was 2.1 versus 2.9 months (HR, 1.39; P = 0.09) with veliparib versus control. Grade 3/4 toxicities were more common with veliparib (69% vs. 58%, P = 0.23). For cancers with HR-DDR defects versus wild-type, median PFS and OS were 7.3 versus 2.5 months (P = 0.05) and 10.1 versus 5.9 months (P = 0.17), respectively, with FOLFIRI, and 2.0 versus 2.1 months (P = 0.62) and 7.4 versus 5.1 months (P = 0.10), respectively, with veliparib plus mFOLFIRI. CONCLUSIONS Veliparib plus mFOLFIRI did not improve survival for metastatic pancreatic cancer. FOLFIRI should be further studied in pancreatic cancers with HR-DDR defects.
Collapse
Affiliation(s)
- E Gabriela Chiorean
- University of Washington School of Medicine, Seattle, Washington.
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Katherine A Guthrie
- Fred Hutchinson Cancer Research Center, Seattle, Washington
- SWOG Statistics and Data Management Center, Seattle, Washington
| | - Philip A Philip
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | | | - Florencia Jalikis
- University of Washington School of Medicine, Seattle, Washington
- Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Michael J Pishvaian
- Georgetown University, Lombardi Cancer Center, Washington, DC
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jordan Berlin
- Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Marcus S Noel
- Georgetown University, Lombardi Cancer Center, Washington, DC
| | | | | | | | - Marc R Radke
- University of Washington School of Medicine, Seattle, Washington
| | - Mai Duong
- Fred Hutchinson Cancer Research Center, Seattle, Washington
- SWOG Statistics and Data Management Center, Seattle, Washington
| | - Shay Bellasea
- Fred Hutchinson Cancer Research Center, Seattle, Washington
- SWOG Statistics and Data Management Center, Seattle, Washington
| | - Andrew M Lowy
- University of California San Diego, Moores Cancer Center, La Jolla, California
| | | |
Collapse
|
12
|
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: 18] [Impact Index Per Article: 4.5] [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
|
13
|
Zhou L, Xiang J, He Y. Research progress on the association between environmental pollutants and the resistance mechanism of PARP inhibitors in ovarian cancer. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:49491-49506. [PMID: 34370190 DOI: 10.1007/s11356-021-15852-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/03/2021] [Indexed: 06/13/2023]
Abstract
The occurrence and progression of ovarian cancer are closely related to genetics and environmental pollutants. Poly(ADP-ribose) polymerase (PARP) inhibitors have been a major breakthrough in the history of ovarian cancer treatment. PARP is an enzyme responsible for post-translational modification of proteins and repair of single-stranded DNA damage. PARP inhibitors can selectively inhibit PARP function, resulting in a synthetic lethal effect on tumor cells defective in homologous recombination repair. However, with large-scale application, drug resistance also inevitably appears. For PARP inhibitors, the diversity and complexity of drug resistance mechanisms have always been difficult problems in clinical treatment. Herein, we mainly summarized the research progress of DNA damage repair and drug resistance mechanisms related to PARP inhibitors and the impact of environmental pollutants on DNA damage repair to aid the development prospects and highlight urgent problems to be solved.
Collapse
Affiliation(s)
- Lina Zhou
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Jiangdong Xiang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Yinyan He
- Department of Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
14
|
Wu HJ, Chu PY. Epigenetic Regulation of Breast Cancer Stem Cells Contributing to Carcinogenesis and Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22158113. [PMID: 34360879 PMCID: PMC8348144 DOI: 10.3390/ijms22158113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Globally, breast cancer has remained the most commonly diagnosed cancer and the leading cause of cancer death among women. Breast cancer is a highly heterogeneous and phenotypically diverse group of diseases, which require different selection of treatments. Breast cancer stem cells (BCSCs), a small subset of cancer cells with stem cell-like properties, play essential roles in breast cancer progression, recurrence, metastasis, chemoresistance and treatments. Epigenetics is defined as inheritable changes in gene expression without alteration in DNA sequence. Epigenetic regulation includes DNA methylation and demethylation, as well as histone modifications. Aberrant epigenetic regulation results in carcinogenesis. In this review, the mechanism of epigenetic regulation involved in carcinogenesis, therapeutic resistance and metastasis of BCSCs will be discussed, and finally, the therapies targeting these biomarkers will be presented.
Collapse
Affiliation(s)
- Hsing-Ju Wu
- Department of Biology, National Changhua University of Education, Changhua 500, Taiwan;
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua 505, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Correspondence: ; Tel.: +886-975611855; Fax: +886-47227116
| |
Collapse
|
15
|
Sun W, Zhang Q, Wang R, Li Y, Sun Y, Yang L. Targeting DNA Damage Repair for Immune Checkpoint Inhibition: Mechanisms and Potential Clinical Applications. Front Oncol 2021; 11:648687. [PMID: 34026622 PMCID: PMC8137908 DOI: 10.3389/fonc.2021.648687] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/30/2021] [Indexed: 12/26/2022] Open
Abstract
DNA damage repair (DDR) pathways play an essential role in maintaining genomic integrity. DDR dysfunction leads to accumulated DNA damage, predisposition to cancer, and high sensitivity to chemotherapy and radiotherapy. Recent studies have demonstrated that DDR status is associated with response to immune checkpoint inhibitors (ICIs). Among the DDR pathways, mismatch repair is one of the most recognized predictive biomarkers for ICIs. Furthermore, preclinical and early clinical studies suggest the rationale of combining agents targeting the DDR pathways, such as poly (ADP-ribose) polymerase (PARP) inhibitors, cyclin-dependent kinase 4/6 (CDK4/6) inhibitors, and ataxia telangiectasia and rad3-related (ATR) kinase inhibitors, with ICIs. In the present review, we describe the predictive role of DDR pathways in ICIs and summarize the advances in potential combination strategies of novel agents targeting DDR with ICIs for cancer treatment.
Collapse
Affiliation(s)
- Wei Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Zhang
- Department of Ultrasonic Diagnosis, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runkun Wang
- Department of Oncology, The First People's hospital of Guangshui, Hubei, China
| | - Yang Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Klein FG, Granier C, Zhao Y, Pan Q, Tong Z, Gschwend JE, Holm PS, Nawroth R. Combination of Talazoparib and Palbociclib as a Potent Treatment Strategy in Bladder Cancer. J Pers Med 2021; 11:jpm11050340. [PMID: 33923231 PMCID: PMC8145096 DOI: 10.3390/jpm11050340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/20/2022] Open
Abstract
The use of cyclin-dependent kinase 4/6 (CDK4/6) inhibitors represents a potent strategy for cancer therapy. Due to the complex molecular network that regulates cell cycle progression, cancer cells often acquire resistance mechanisms against these inhibitors. Previously, our group identified molecular factors conferring resistance to CDK4/6 inhibition in bladder cancer (BLCA) that also included components within the DNA repair pathway. In this study, we validated whether a combinatory treatment approach of the CDK4/6 inhibitor Palbociclib with Poly-(ADP-Ribose) Polymerase (PARP) inhibitors improves therapy response in BLCA. First, a comparison of PARP inhibitors Talazoparib and Olaparib showed superior efficacy of Talazoparib in vitro and displayed high antitumor activity in xenografts in the chicken chorioallantoic membrane (CAM) model. Moreover, the combination of Talazoparib and the CDK4/6 inhibitor Palbociclib synergistically reduced tumor growth in Retinoblastoma protein (RB)-positive BLCA in vitro and in a CAM model, an effect that relies on Palbociclib-induced cell cycle arrest in G0/G1-phase complemented by a G2 arrest induced by Talazoparib. Interestingly, Talazoparib-induced apoptosis was reduced by Palbociclib. The combination of Palbociclib and Talazoparib effectively enhances BLCA therapy, and RB is a molecular biomarker of response to this treatment regimen.
Collapse
Affiliation(s)
- Florian G. Klein
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany; (F.G.K.); (C.G.); (Y.Z.); (Q.P.); (Z.T.); (J.E.G.); (P.S.H.)
| | - Charlène Granier
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany; (F.G.K.); (C.G.); (Y.Z.); (Q.P.); (Z.T.); (J.E.G.); (P.S.H.)
| | - Yuling Zhao
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany; (F.G.K.); (C.G.); (Y.Z.); (Q.P.); (Z.T.); (J.E.G.); (P.S.H.)
| | - Qi Pan
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany; (F.G.K.); (C.G.); (Y.Z.); (Q.P.); (Z.T.); (J.E.G.); (P.S.H.)
| | - Zhichao Tong
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany; (F.G.K.); (C.G.); (Y.Z.); (Q.P.); (Z.T.); (J.E.G.); (P.S.H.)
| | - Jürgen E. Gschwend
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany; (F.G.K.); (C.G.); (Y.Z.); (Q.P.); (Z.T.); (J.E.G.); (P.S.H.)
| | - Per Sonne Holm
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany; (F.G.K.); (C.G.); (Y.Z.); (Q.P.); (Z.T.); (J.E.G.); (P.S.H.)
- Department of Oral and Maxillofacial Surgery, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Roman Nawroth
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany; (F.G.K.); (C.G.); (Y.Z.); (Q.P.); (Z.T.); (J.E.G.); (P.S.H.)
- Correspondence: ; Tel.: +49-89-41402553
| |
Collapse
|
17
|
Molecular mechanisms of breast cancer chemoresistance by immune checkpoints. Life Sci 2020; 263:118604. [PMID: 33096117 DOI: 10.1016/j.lfs.2020.118604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 01/02/2023]
Abstract
Progression of resistance to chemotherapy in breast cancer (BC) has been recognized as a main factor in decreasing the survival of patients with this malignancy. Recent investigations have described the involvement of immune checkpoint molecules in the progress of drug resistance in breast carcinoma patients. In the present study, the molecular participation of immune checkpoint factors in chemoresistance of BC both in-vitro and in-vivo is reviewed. Numerous immune checkpoints such as PD-1/PD-L1, CTLA-4, B7-H3, B7-H4, B7-1, and B7-2 have been specified as positive regulators of resistance to various drug types in BC. In several molecular pathways of drug resistance in BC, immune checkpoints affect the chemoresistance of this cancer in a drug- and cell-type-dependent manner. In addition, immune checkpoints promote chemoresistance in response to particular drugs in specific BC cell lines. Furthermore, several the immune checkpoint molecules have not been evaluated in the field of the chemoresistance in breast malignancy either in-vitro or in-vivo. Overall, investigations have indicated that targeting immune checkpoint molecules may be considered as a novel method to improve existing anti-BC treatments.
Collapse
|
18
|
Corrales-Sánchez V, Noblejas-López MDM, Nieto-Jiménez C, Pérez-Peña J, Montero JC, Burgos M, Galán-Moya EM, Pandiella A, Ocaña A. Pharmacological screening and transcriptomic functional analyses identify a synergistic interaction between dasatinib and olaparib in triple-negative breast cancer. J Cell Mol Med 2020; 24:3117-3127. [PMID: 32032474 PMCID: PMC7077558 DOI: 10.1111/jcmm.14980] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 10/29/2019] [Accepted: 12/09/2019] [Indexed: 12/18/2022] Open
Abstract
Identification of druggable vulnerabilities is a main objective in triple‐negative breast cancer (TNBC), where no curative therapies exist. Gene set enrichment analyses (GSEA) and a pharmacological evaluation using a library of compounds were used to select potential druggable combinations. MTT and studies with semi‐solid media were performed to explore the activity of the combinations. TNBC cell lines (MDAMB‐231, BT549, HS‐578T and HCC3153) and an additional panel of 16 cell lines were used to assess the activity of the two compounds. Flow cytometry experiments and biochemical studies were also performed to explore the mechanism of action. GSEA were performed using several data sets (GSE21422, GSE26910, GSE3744, GSE65194 and GSE42568), and more than 35 compounds against the identified functions were evaluated to discover druggable opportunities. Analyses done with the Chou and Talalay algorithm confirmed the synergy of dasatinib and olaparib. The combination of both agents significantly induced apoptosis in a caspase‐dependent manner and revealed a pleotropic effect on cell cycle: Dasatinib arrested cells in G0/G1 and olaparib in G2/M. Dasatinib inhibited pChk1 and induced DNA damage measured by pH2AX, and olaparib increased pH3. Finally, the effect of the combination was also evaluated in a panel of 18 cell lines representative of the most frequent solid tumours, observing a particularly synergism in ovarian cancer. Breast cancer, triple negative, dasatinib, olaparib, screening.
Collapse
Affiliation(s)
| | - María Del Mar Noblejas-López
- Translational Research Unit, Albacete University Hospital, Albacete, Spain.,Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha, Albacete, Spain
| | - Cristina Nieto-Jiménez
- Translational Research Unit, Albacete University Hospital, Albacete, Spain.,Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha, Albacete, Spain
| | - Javier Pérez-Peña
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha, Albacete, Spain
| | - Juan Carlos Montero
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Salamanca, Spain.,IBSAL, Salamanca, Spain.,CIBERONC, Salamanca, Spain
| | - Miguel Burgos
- Translational Research Unit, Albacete University Hospital, Albacete, Spain
| | - Eva M Galán-Moya
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha, Albacete, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Salamanca, Spain.,IBSAL, Salamanca, Spain.,CIBERONC, Salamanca, Spain.,CSIC, Salamanca, Spain
| | - Alberto Ocaña
- Translational Research Unit, Albacete University Hospital, Albacete, Spain.,Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha, Albacete, Spain.,Unidad de nuevas terapias y Oncología traslacional, IDISSC and CIBERONC, Hospital Clínico Universitario San Carlos, Madrid, Spain
| |
Collapse
|
19
|
Hu G, Tu W, Yang L, Peng G, Yang L. ARID1A deficiency and immune checkpoint blockade therapy: From mechanisms to clinical application. Cancer Lett 2020; 473:148-155. [PMID: 31911080 DOI: 10.1016/j.canlet.2020.01.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/15/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023]
Abstract
The AT-rich interaction domain 1A (ARID1A, also known as BAF250a) is a chromatin remodeling gene, which frequently mutates across a broad spectrum of cancers with loss expression of the ARID1A protein. Recently, the association between ARID1A deficiency and immune checkpoint blockade (ICB) therapy has been reported. ARID1A deficiency contributes to the high microsatellite instability phenotype, increases tumor mutation burden, elevates expression of programmed cell death ligand 1 (PD-L1), and modulates the immune microenvironment, supporting the view that ARID1A loss might serve as a predictive biomarker for ICB. Furthermore, the therapeutic targeting strategies, which show "synthetic lethality" with ARID1A deficiency, exhibit potential synergy with ICB. We collectively reviewed the mechanisms underlying the correlation between ARID1A deficiency and ICB, the predictive function of ARID1A deficiency for ICB, and potential combined strategies of targeting agents, vulnerable for ARID1A deficiency, with ICB in cancer treatment.
Collapse
Affiliation(s)
- Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wei Tu
- Department of Rheumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Liu Yang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Guang Peng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Lin Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
20
|
Raineri A, Prodomini S, Fasoli S, Gotte G, Menegazzi M. Influence of onconase in the therapeutic potential of PARP inhibitors in A375 malignant melanoma cells. Biochem Pharmacol 2019; 167:173-181. [PMID: 31185226 DOI: 10.1016/j.bcp.2019.06.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 06/06/2019] [Indexed: 12/12/2022]
Abstract
Human malignant melanoma is one of the most aggressive cancers, accompanied with poor prognosis, metastatic evolution and high mortality. Many strategies have been developed using BRAF and MEK inhibitors in spite of the classic therapy with alkylating agents, but failure related to the ability of the tumor to activate alternative proliferation pathways occurred after promising initial successes. Poly(ADP-ribose) polymerase (PARP) enzymes are well known to be crucial for DNA damage response, and PARP inhibition results in the accumulation of DNA strand breaks that induce cell injury. For this reason, PARP-inhibitors (PARPi) have become promising tools to counteract many cancer types. One of the most used by clinicians is olaparib, that, however, showed again cancer resistance in patients. Thus, new generation molecules have been designed mainly to counteract this problem. Among them, we chose to test AZD2461 on the particularly aggressive human melanoma A375 cell line. This drug is a PARPi significantly less prone than olaparib to undergo the P-glycoprotein-mediated efflux mechanism, one of those responsible for resistance, that in turn is the main adversity in melanoma therapy. Then, we analysed AZD2461 also together with the enzyme onconase (ONC) on the same A375 cells, to investigate if the combination of drugs could possibly increase the in vitro antitumor activity. ONC is a small amphibian "pancreatic-type" ribonuclease that is able to exert a remarkable antitumor activity against many cancers, either in vitro or in vivo, principally because it can evade the ubiquitous ribonuclease cytosolic inhibitor thanks to its structural determinants. Hence, ONC became relevant in the use of protein-drug strategies against incurable cancers. The studies performed in this work showed that both drugs definitely affect A375 cells viability by inducing cytostatic and pro-apoptotic effects in a time- and dose-dependent manner, either if administered alone or in combination. Although we registered low synergistic effects with the combination of the two drugs, we found that AZD2461 did not induce resistance in A375 after two months treatment with high concentration of this molecule. Moreover, we underline that A375 cells treated for a prolonged time with AZD2461 were definitely more susceptible than parental A375 cells to the pro-apoptotic action of ONC. Considering also the different inhibitory effects of the two drugs on TNF-α gene expression and NF-κB DNA-binding, the tuning of their combined delivery to the A375 tumor cell line might open a promising scenario for future therapeutic applications devoted to defeat human melanoma.
Collapse
Affiliation(s)
- Alice Raineri
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Sara Prodomini
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Sabrina Fasoli
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Giovanni Gotte
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Marta Menegazzi
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy.
| |
Collapse
|
21
|
Investigating circulating tumor cells and distant metastases in patient-derived orthotopic xenograft models of triple-negative breast cancer. Breast Cancer Res 2019; 21:98. [PMID: 31462307 PMCID: PMC6714238 DOI: 10.1186/s13058-019-1182-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) represent a temporal "snapshot" of a patient's cancer and changes that occur during disease evolution. There is an extensive literature studying CTCs in breast cancer patients, and particularly in those with metastatic disease. In parallel, there is an increasing use of patient-derived models in preclinical investigations of human cancers. Yet studies are still limited demonstrating CTC shedding and metastasis formation in patient-derived models of breast cancer. METHODS We used seven patient-derived orthotopic xenograft (PDOX) models generated from triple-negative breast cancer (TNBC) patients to study CTCs and distant metastases. Tumor fragments from PDOX tissue from each of the seven models were implanted into 57 NOD scid gamma (NSG) mice, and tumor growth and volume were monitored. Human CTC capture from mouse blood was first optimized on the marker-agnostic Vortex CTC isolation platform, and whole blood was processed from 37 PDOX tumor-bearing mice. RESULTS Staining and imaging revealed the presence of CTCs in 32/37 (86%). The total number of CTCs varied between different PDOX tumor models and between individual mice bearing the same PDOX tumors. CTCs were heterogeneous and showed cytokeratin (CK) positive, vimentin (VIM) positive, and mixed CK/VIM phenotypes. Metastases were detected in the lung (20/57, 35%), liver (7/57, 12%), and brain (1/57, less than 2%). The seven different PDOX tumor models displayed varying degrees of metastatic potential, including one TNBC PDOX tumor model that failed to generate any detectable metastases (0/8 mice) despite having CTCs present in the blood of 5/5 tested, suggesting that CTCs from this particular PDOX tumor model may typify metastatic inefficiency. CONCLUSION PDOX tumor models that shed CTCs and develop distant metastases represent an important tool for investigating TNBC.
Collapse
|
22
|
Przybycinski J, Nalewajska M, Marchelek-Mysliwiec M, Dziedziejko V, Pawlik A. Poly-ADP-ribose polymerases (PARPs) as a therapeutic target in the treatment of selected cancers. Expert Opin Ther Targets 2019; 23:773-785. [PMID: 31394942 DOI: 10.1080/14728222.2019.1654458] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The implementation of poly-ADP-ribose polymerase (PARP) inhibitors for therapy has created potential treatments for a wide spectrum of malignancies involving DNA damage repair gene abnormalities. PARPs are a group of enzymes that are responsible for detecting and repairing DNA damage and therefore play a key role in maintaining cell function and integrity. PARP inhibitors are drugs that target DNA repair deficiencies. Inhibiting PARP activity in cancer cells causes cell death. Areas covered: This review summarizes the role of PARP inhibitors in the treatment of cancer. We performed a systematic literature search in February 2019 in the electronic databases PubMed and EMBASE. Our search terms were the following: PARP, PARP inhibitors, PARPi, Poly ADP ribose polymerase, cancer treatment. We discuss PARP inhibitors currently being investigated in cancer clinical trials, their safety profiles, clinical resistance, combined therapeutic approaches and future challenges. Expert Opinion: The future could bring novel PARP inhibitors with greater DNA trapping potential, better safety profiles and improved combined therapies involving hormonal, chemo-, radio- or immunotherapies. Progress may afford wider indications for PARP inhibitors in the treatment of cancer and the utilization for cancer prevention in high-risk mutation carriers. Research efforts should focus on identifying novel drugs that target DNA repair deficiencies.
Collapse
Affiliation(s)
- Jarosław Przybycinski
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University , Szczecin , Poland
| | - Magdalena Nalewajska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University , Szczecin , Poland
| | | | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University , Szczecin , Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University , Szczecin , Poland
| |
Collapse
|
23
|
Mechanisms of Genomic Instability in Breast Cancer. Trends Mol Med 2019; 25:595-611. [DOI: 10.1016/j.molmed.2019.04.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 12/22/2022]
|
24
|
Wu F, McCuaig RD, Sutton CR, Tan AHY, Jeelall Y, Bean EG, Dai J, Prasanna T, Batham J, Malik L, Yip D, Dahlstrom JE, Rao S. Nuclear-Biased DUSP6 Expression is Associated with Cancer Spreading Including Brain Metastasis in Triple-Negative Breast Cancer. Int J Mol Sci 2019; 20:3080. [PMID: 31238530 PMCID: PMC6627630 DOI: 10.3390/ijms20123080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/20/2019] [Accepted: 06/22/2019] [Indexed: 12/18/2022] Open
Abstract
DUSP6 is a dual-specificity phosphatase (DUSP) involved in breast cancer progression, recurrence, and metastasis. DUSP6 is predominantly cytoplasmic in HER2+ primary breast cancer cells, but the expression and subcellular localization of DUSPs, especially DUSP6, in HER2-positive circulating tumor cells (CTCs) is unknown. Here we used the DEPArray system to identify and isolate CTCs from metastatic triple negative breast cancer (TNBC) patients and performed single-cell NanoString analysis to quantify cancer pathway gene expression in HER2-positive and HER2-negative CTC populations. All TNBC patients contained HER2-positive CTCs. HER2-positive CTCs were associated with increased ERK1/ERK2 expression, which are direct DUSP6 targets. DUSP6 protein expression was predominantly nuclear in breast CTCs and the brain metastases but not pleura or lung metastases of TNBC patients. Therefore, nuclear DUSP6 may play a role in the association with cancer spreading in TNBC patients, including brain metastasis.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents, Immunological/pharmacology
- Biomarkers, Tumor
- Brain Neoplasms/secondary
- Cell Line, Tumor
- Cell Nucleus/genetics
- Disease Models, Animal
- Dual Specificity Phosphatase 6/antagonists & inhibitors
- Dual Specificity Phosphatase 6/genetics
- Dual Specificity Phosphatase 6/metabolism
- Female
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic/drug effects
- Histones/metabolism
- Humans
- MAP Kinase Signaling System
- Mice
- Neoplasm Invasiveness
- Neoplasm Staging
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Protein Binding
- Protein Transport
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Single-Cell Analysis
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/therapy
- Xenograft Model Antitumor Assays
- p300-CBP Transcription Factors/metabolism
Collapse
Affiliation(s)
- Fan Wu
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Robert D McCuaig
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Christopher R Sutton
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Abel H Y Tan
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Yoshni Jeelall
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Elaine G Bean
- Anatomical Pathology, ACT Pathology, The Canberra Hospital, Canberra health Services, Garran ACT 2606, Australia.
| | - Jin Dai
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Thiru Prasanna
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
- Department of Medical Oncology, The Canberra Hospital, Canberra health Services, Garran ACT 2606, Australia.
| | - Jacob Batham
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Laeeq Malik
- Department of Medical Oncology, The Canberra Hospital, Canberra health Services, Garran ACT 2606, Australia.
- ANU Medical School, College of Health and Medicine, The Australian National University, Canberra ACT 0200, Australia.
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Canberra health Services, Garran ACT 2606, Australia.
- ANU Medical School, College of Health and Medicine, The Australian National University, Canberra ACT 0200, Australia.
| | - Jane E Dahlstrom
- Anatomical Pathology, ACT Pathology, The Canberra Hospital, Canberra health Services, Garran ACT 2606, Australia.
- ANU Medical School, College of Health and Medicine, The Australian National University, Canberra ACT 0200, Australia.
| | - Sudha Rao
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| |
Collapse
|
25
|
Yonemori K, Shimomura A, Yasojima H, Masuda N, Aogi K, Takahashi M, Naito Y, Shimizu S, Nakamura R, Hashimoto J, Yamamoto H, Hirakawa A, Michimae H, Hamada A, Yoshida T, Sukigara T, Tamura K, Fujiwara Y. A phase I/II trial of olaparib tablet in combination with eribulin in Japanese patients with advanced or metastatic triple-negative breast cancer previously treated with anthracyclines and taxanes. Eur J Cancer 2019; 109:84-91. [DOI: 10.1016/j.ejca.2018.11.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/09/2018] [Accepted: 11/10/2018] [Indexed: 01/17/2023]
|
26
|
Criscuolo D, Morra F, Giannella R, Visconti R, Cerrato A, Celetti A. New combinatorial strategies to improve the PARP inhibitors efficacy in the urothelial bladder Cancer treatment. J Exp Clin Cancer Res 2019; 38:91. [PMID: 30791940 PMCID: PMC6385418 DOI: 10.1186/s13046-019-1089-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/06/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Novel therapeutic strategies are urgently needed for the treatment of metastatic Urothelial Bladder Cancer. DNA damaging repair (DDR) targeting has been introduced in cinical trials for bladder cancer patients that carry alterations in homologous DNA repair genes, letting to envisage susceptibility to the Poly (adenosine diphosphate [ADP]) ribose polymerase (PARP) inhibitors. MAIN BODY PARP inhibition, by amplifying the DNA damage, augments the mutational burden and promotes the immune priming of the tumor by increasing the neoantigen exposure and determining upregulation of programmed death ligand 1 (PD-L1) expression. Thus, the combination of PARP-inhibition and the PD/PD-L1 targeting may represent a compelling strategy to treat bladder cancer and has been introduced in recent clinical trials. The targeting of DDR has been also used in combination with epigenetic drugs able to modulate the expression of genes involved in DDR, and also able to act as immunomodulator agents suggesting their use in combination with immune-checkpoint inhibitors. CONCLUSION In conclusion, it may be envisaged the combination of three classes of drugs to treat bladder cancer, by targeting the DDR process in a tumor context of DDR defect, together with epigenetic agents and immune-checkpoint inhibitors, whose association may amplify the effects and reduce the doses and the toxicity of each single drug.
Collapse
Affiliation(s)
- Daniela Criscuolo
- Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, Naples, Italy
| | - Francesco Morra
- Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, Naples, Italy
| | | | - Roberta Visconti
- Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, Naples, Italy
| | - Aniello Cerrato
- Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, Naples, Italy
| | - Angela Celetti
- Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, Naples, Italy
| |
Collapse
|
27
|
Kalimutho M, Nones K, Srihari S, Duijf PHG, Waddell N, Khanna KK. Patterns of Genomic Instability in Breast Cancer. Trends Pharmacol Sci 2019; 40:198-211. [PMID: 30736983 DOI: 10.1016/j.tips.2019.01.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/14/2018] [Accepted: 01/08/2019] [Indexed: 01/02/2023]
Abstract
Breast cancer is one of the most common cancers affecting women. Despite significant improvements in overall survival, it remains a significant cause of death worldwide. Genomic instability (GI) is a hallmark of cancer and plays a pivotal role in breast cancer development and progression. In the past decade, high-throughput technologies have provided a wealth of information that has facilitated the identification of a diverse repertoire of mutated genes and mutational processes operative across cancers. Here, we review recent findings on genomic alterations and mutational processes in breast cancer pathogenesis. Most importantly, we summarize the clinical challenges and opportunities to utilize omics-based signatures for better management of breast cancer patients and treatment decision-making.
Collapse
Affiliation(s)
- Murugan Kalimutho
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD 4006, Australia.
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD 4006, Australia
| | - Sriganesh Srihari
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Brisbane, QLD 4102, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD 4006, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD 4006, Australia.
| |
Collapse
|
28
|
Prasanna T, Wu F, Khanna KK, Yip D, Malik L, Dahlstrom JE, Rao S. Optimizing poly (ADP-ribose) polymerase inhibition through combined epigenetic and immunotherapy. Cancer Sci 2018; 109:3383-3392. [PMID: 30230653 PMCID: PMC6215877 DOI: 10.1111/cas.13799] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 09/05/2018] [Accepted: 09/09/2018] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with poor survival outcomes. Currently, there are no targeted therapies available for TNBCs despite remarkable progress in targeted and immune-directed therapies for other solid organ malignancies. Poly (ADP-ribose) polymerase inhibitors (PARPi) are effective anticancer drugs that produce good initial clinical responses, especially in homologous recombination DNA repair-deficient cancers. However, resistance is the rule rather than the exception, and recurrent tumors tend to have an aggressive phenotype associated with poor survival. Many efforts have been made to overcome PARPi resistance, mostly by targeting genes and effector proteins participating in homologous recombination that are overexpressed during PARPi therapy. Due to many known and unknown compensatory pathways, genes, and effector proteins, overlap and shared resistance are common. Overexpression of programmed cell death-ligand 1 (PD-L1) and cancer stem cell (CSC) sparing are novel PARPi resistance hypotheses. Although adding programmed cell death-1 (PD-1)/PD-L1 inhibitors to PARPi might improve immunogenic cell death and be crucial for durable responses, they are less likely to target the CSC population that drives recurrent tumor growth. Lysine-specific histone demethylase-1A and histone deacetylase inhibitors have shown promising activity against CSCs. Combining epigenetic drugs such as lysine-specific histone demethylase-1A inhibitors or histone deacetylase inhibitors with PARPi/anti-PD-1/PD-L1 is a novel, potentially synergistic strategy for priming tumors and overcoming resistance. Furthermore, such an approach could pave the way for the identification of new upstream epigenetic and genetic signatures.
Collapse
Affiliation(s)
- Thiru Prasanna
- Health Research InstituteFaculty of ESTeMUniversity of CanberraCanberraACTAustralia
- Department of Medical OncologyThe Canberra HospitalCanberraACTAustralia
| | - Fan Wu
- Health Research InstituteFaculty of ESTeMUniversity of CanberraCanberraACTAustralia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Desmond Yip
- Department of Medical OncologyThe Canberra HospitalCanberraACTAustralia
- ANU Medical SchoolAustralian National UniversityCanberraACTAustralia
| | - Laeeq Malik
- Department of Medical OncologyThe Canberra HospitalCanberraACTAustralia
- ANU Medical SchoolAustralian National UniversityCanberraACTAustralia
| | - Jane E. Dahlstrom
- ANU Medical SchoolAustralian National UniversityCanberraACTAustralia
- Department of Anatomical PathologyACT PathologyThe Canberra HospitalCanberraACTAustralia
| | - Sudha Rao
- Health Research InstituteFaculty of ESTeMUniversity of CanberraCanberraACTAustralia
| |
Collapse
|