1
|
Kieronska-Rudek A, Zuhra K, Ascenção K, Chlopicki S, Szabo C. The PARP inhibitor olaparib promotes senescence in murine macrophages. GeroScience 2025:10.1007/s11357-025-01679-6. [PMID: 40327273 DOI: 10.1007/s11357-025-01679-6] [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: 04/01/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
Cellular senescence is a multifaceted process involving cell cycle arrest, telomere shortening, and the accumulation of DNA damage associated with aging and cellular stress. It is marked by persistent cell cycle arrest and DNA damage accumulation, and plays an increasingly recognized role in age-related diseases and cancer therapy. Olaparib, a poly (ADP-ribose) polymerase (PARP) inhibitor, is approved for use in ovarian cancer treatment. We hypothesized that olaparib may influence senescence by inhibiting DNA damage repair, and investigated its effects on non-senescent and replicatively senescent murine macrophages (RAW 264.7 cells). Senescent cells exhibited elevated baseline levels of PARP1 expression, PARylation, and DNA damage relative to non-senescent control cells. Olaparib amplified these differences by upregulating senescence markers (SA-β-gal and p21), inhibiting proliferation, and exacerbating DNA damage. Many of its effects were more pronounced in senescent cells. At higher concentrations (10-30 µM), olaparib induced significant cytotoxicity through mixed apoptotic and necrotic mechanisms, with senescent cells exhibiting a predominantly necrotic response. Interestingly, both mitochondrial activity and cellular bioenergetics were elevated in senescent cells at baseline, and were more severely impaired by olaparib compared to non-senescent control cells. These findings underscore olaparib's enhanced cytotoxic and pro-senescent effects in senescent immune cells and suggest potential implications for its use in elderly cancer patients with an increased burden of senescent cells.
Collapse
Affiliation(s)
- Anna Kieronska-Rudek
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Karim Zuhra
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Kelly Ascenção
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Csaba Szabo
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
2
|
Zarco N, Dovas A, de Araujo Farias V, Nagaiah NK, Haddock A, Sims PA, Hambardzumyan D, Meyer CT, Canoll P, Rosenfeld SS, Kenchappa RS. Resistance to spindle inhibitors in glioblastoma depends on STAT3 and therapy induced senescence. iScience 2024; 27:111311. [PMID: 39640583 PMCID: PMC11617384 DOI: 10.1016/j.isci.2024.111311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/16/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
While mitotic spindle inhibitors specifically kill proliferating tumor cells without the toxicities of microtubule poisons, resistance has limited their clinical utility. Treating glioblastomas with the spindle inhibitors ispinesib, alisertib, or volasertib creates a subpopulation of therapy induced senescent cells that resist these drugs by relying upon the anti-apoptotic and metabolic effects of activated STAT3. Furthermore, these senescent cells expand the repertoire of cells resistant to these drugs by secreting an array of factors, including TGFβ, which induce proliferating cells to exit mitosis and become quiescent-a state that also resists spindle inhibitors. Targeting STAT3 restores sensitivity to each of these drugs by depleting the senescent subpopulation and inducing quiescent cells to enter the mitotic cycle. These results support a therapeutic strategy of targeting STAT3-dependent therapy-induced senescence to enhance the efficacy of spindle inhibitors for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Natanael Zarco
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | | | - Ashley Haddock
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Dolores Hambardzumyan
- Departments of Oncological Sciences and Neurosurgery, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Steven S. Rosenfeld
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rajappa S. Kenchappa
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
3
|
Malik U, Pal D. Isoxazole compounds: Unveiling the synthetic strategy, in-silico SAR & toxicity studies and future perspective as PARP inhibitor in cancer therapy. Eur J Med Chem 2024; 279:116898. [PMID: 39353240 DOI: 10.1016/j.ejmech.2024.116898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Latest developments in cancer treatment have shed a light on the crucial role of PARP inhibitors that enhance the treatment effectiveness by modifying abnormal repair pathways. PARP inhibitors, such as Olaparib, Rucaparib, Niraparib, and Talazoparib have been approved in a number of cancers including BRCA 1/BRCA2 associated malignancies although there are many difficulties as therapeutical resistance. Besides the conventional synthetic drugs, natural compounds such as flavones and flavonoids have been found to be PARP inhibitors but only in preclinical studies. Isoxazole is very important class of potential candidates for medicinal chemistry with anti-cancer and other pharmacological activities. At present, there are no approved PARP inhibitors of isoxazole origin but their ability to hit many pathways inside the cancer cells points out on its importance for future treatments design. In drug development, isoxazoles are helpful because of the molecular design flexibility that may be enhanced using various synthetic approaches. This review highlights the molecular mechanisms of PARP inhibition, importance of isoxazole compounds and present advances in their synthetic strategies that demonstrate promise for these agents as new anticancer drugs. It emphasizes that isoxazole-based PARP inhibitors compounds could be novel anti-cancer drugs. Through this review, we hope to grow a curiosity in additional explorations of isoxazole-based PARP inhibitors and their applications in the trends of novel insights towards precision cancer therapy.
Collapse
Affiliation(s)
- Udita Malik
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G., 495009, India
| | - Dilipkumar Pal
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G., 495009, India.
| |
Collapse
|
4
|
Zarco N, Dovas A, de Araujo Farias V, Nagaiah NK, Haddock A, Sims PA, Hambardzumyan D, Meyer CT, Canoll P, Rosenfeld SS, Kenchappa RS. Resistance to Spindle Inhibitors in Glioblastoma Depends on STAT3 and Therapy Induced Senescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598115. [PMID: 38895402 PMCID: PMC11185785 DOI: 10.1101/2024.06.09.598115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
While mitotic spindle inhibitors specifically kill proliferating tumor cells without the toxicities of microtubule poisons, resistance has limited their clinical utility. Treating glioblastomas with the spindle inhibitors ispinesib, alisertib, or volasertib creates a subpopulation of therapy induced senescent cells that resist these drugs by relying upon the anti-apoptotic and metabolic effects of activated STAT3. Furthermore, these senescent cells expand the repertoire of cells resistant to these drugs by secreting an array of factors, including TGFβ, which induce proliferating cells to exit mitosis and become quiescent-a state that also resists spindle inhibitors. Targeting STAT3 restores sensitivity to each of these drugs by depleting the senescent subpopulation and inducing quiescent cells to enter the mitotic cycle. These results support a therapeutic strategy of targeting STAT3-dependent therapy-induced senescence to enhance the efficacy of spindle inhibitors for the treatment of glioblastoma. Highlights • Resistance to non-microtubule spindle inhibitors limits their efficacy in glioblastoma and depends on STAT3.• Resistance goes hand in hand with development of therapy induced senescence (TIS).• Spindle inhibitor resistant glioblastomas consist of three cell subpopulations-proliferative, quiescent, and TIS-with proliferative cells sensitive and quiescent and TIS cells resistant.• TIS cells secrete TGFβ, which induces proliferative cells to become quiescent, thereby expanding the population of resistant cells in a spindle inhibitor resistant glioblastoma• Treatment with a STAT3 inhibitor kills TIS cells and restores sensitivity to spindle inhibitors.
Collapse
|
5
|
Tóth F, Moftakhar Z, Sotgia F, Lisanti MP. In Vitro Investigation of Therapy-Induced Senescence and Senescence Escape in Breast Cancer Cells Using Novel Flow Cytometry-Based Methods. Cells 2024; 13:841. [PMID: 38786063 PMCID: PMC11120107 DOI: 10.3390/cells13100841] [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: 04/12/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Although cellular senescence was originally defined as an irreversible form of cell cycle arrest, in therapy-induced senescence models, the emergence of proliferative senescence-escaped cancer cells has been reported by several groups, challenging the definition of senescence. Indeed, senescence-escaped cancer cells may contribute to resistance to cancer treatment. Here, to study senescence escape and isolate senescence-escaped cells, we developed novel flow cytometry-based methods using the proliferation marker Ki-67 and CellTrace CFSE live-staining. We investigated the role of a novel senescence marker (DPP4/CD26) and a senolytic drug (azithromycin) on the senescence-escaping ability of MCF-7 and MDA-MB-231 breast cancer cells. Our results show that the expression of DPP4/CD26 is significantly increased in both senescent MCF-7 and MDA-MB-231 cells. While not essential for senescence induction, DPP4/CD26 contributed to promoting senescence escape in MCF-7 cells but not in MDA-MB-231 cells. Our results also confirmed the potential senolytic effect of azithromycin in senescent cancer cells. Importantly, the combination of azithromycin and a DPP4 inhibitor (sitagliptin) demonstrated a synergistic effect in senescent MCF-7 cells and reduced the number of senescence-escaped cells. Although further research is needed, our results and novel methods could contribute to the investigation of the mechanisms of senescence escape and the identification of potential therapeutic targets. Indeed, DPP4/CD26 could be a promising marker and a novel target to potentially decrease senescence escape in cancer.
Collapse
Affiliation(s)
- Fanni Tóth
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
- The CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Wien, Vienna, Austria
| | - Zahra Moftakhar
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| | - Federica Sotgia
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| | - Michael P. Lisanti
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| |
Collapse
|
6
|
Saleh T. Therapy-induced senescence is finally escapable, what is next? Cell Cycle 2024; 23:713-721. [PMID: 38879812 PMCID: PMC11229739 DOI: 10.1080/15384101.2024.2364579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024] Open
Abstract
Several breakthrough articles have recently confirmed the ability of tumor cells to escape the stable cell cycle arrest imposed by Therapy-Induced Senescence (TIS). Subsequently, accepting the hypothesis that TIS is escapable should encourage serious reassessments of the fundamental roles of senescence in cancer treatment. The potential for escape from TIS undermines the well-established tumor suppressor function of senescence, proposes it as a mechanism of tumor dormancy leading to disease recurrence and invites for further investigation of its unfavorable contribution to cancer therapy outcomes. Moreover, escaping TIS strongly indicates that the elimination of senescent tumor cells, primarily through pharmacological means, is a suitable approach for increasing the efficacy of cancer treatment, one that still requires further exploration. This commentary provides an overview of the recent evidence that unequivocally demonstrated the ability of therapy-induced senescent tumor cells in overcoming the terminal growth arrest fate and provides future perspectives on the roles of TIS in tumor biology.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
7
|
Zhang Y, Liang L, Li Z, Huang Y, Jiang M, Zou B, Xu Y. Polyadenosine diphosphate-ribose polymerase inhibitors: advances, implications, and challenges in tumor radiotherapy sensitization. Front Oncol 2023; 13:1295579. [PMID: 38111536 PMCID: PMC10726039 DOI: 10.3389/fonc.2023.1295579] [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: 09/16/2023] [Accepted: 11/22/2023] [Indexed: 12/20/2023] Open
Abstract
Polyadenosine diphosphate-ribose polymerase (PARP) is a key modifying enzyme in cells, which participates in single-strand break repair and indirectly affects double-strand break repair. PARP inhibitors have shown great potential in oncotherapy by exploiting DNA damage repair pathways, and several small molecule PARP inhibitors have been approved by the U.S. Food and Drug Administration for treating various tumor types. PARP inhibitors not only have significant antitumor effects but also have some synergistic effects when combined with radiotherapy; therefore they have potential as radiation sensitizers. Here, we reviewed the advances and implications of PARP inhibitors in tumor radiotherapy sensitization. First, we summarized the multiple functions of PARP and the mechanisms by which its inhibitors exert antitumor effects. Next, we discuss the immunomodulatory effects of PARP and its inhibitors in tumors. Then, we described the theoretical basis of using PARP inhibitors in combination with radiotherapy and outlined their importance in oncological radiotherapy. Finally, we reviewed the current challenges in this field and elaborated on the future applications of PARP inhibitors as radiation sensitizers. A comprehensive understanding of the mechanism, optimal dosing, long-term safety, and identification of responsive biomarkers remain key challenges to integrating PARP inhibition into the radiotherapy management of cancer patients. Therefore, extensive research in these areas would facilitate the development of precision radiotherapy using PARP inhibitors to improve patient outcomes.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lijie Liang
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Li
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Huang
- College of Management, Sichuan Agricultural University, Chengdu, China
| | - Ming Jiang
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Song KX, Wang JX, Huang D. Therapy-induced senescent tumor cells in cancer relapse. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:273-278. [PMID: 39036667 PMCID: PMC11256611 DOI: 10.1016/j.jncc.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 07/23/2024] Open
Abstract
Cellular senescence is characterized by a generally irreversible cell cycle arrest and the secretion of bioactive factors known as the senescence-associated secretory phenotype (SASP). In an oncogenic context, senescence is considered a tumor suppressive mechanism as it prevents cell proliferation and inhibits the progression from pre-malignant to malignant disease. However, recent studies have demonstrated that senescent tumor cells, which could spontaneously exist within cancer tissues or arise in response to various cancer interventions (the so-called therapy-induced senescence, TIS), can acquire pro-tumorigenic properties and are capable of driving local and metastatic relapse. This highlights the complex and multifaceted nature of cellular senescence in cancer biology. Here, we summarize the current knowledge of the pathological function of therapy-induced senescent tumor cells and discuss possible mechanisms by which tumor cell senescence contributes to cancer relapse. We also discuss implications for future studies toward targeting these less appreciated cells.
Collapse
Affiliation(s)
- Ke-Xin Song
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jun-Xian Wang
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - De Huang
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
9
|
Softah A, Alotaibi MR, Alhoshani AR, Saleh T, Alhazzani K, Almutairi MM, AlRowis R, Alshehri S, Albekairy NA, Harada H, Boyd R, Chakraborty E, Gewirtz DA, As Sobeai HM. The Combination of Radiation with PARP Inhibition Enhances Senescence and Sensitivity to the Senolytic, Navitoclax, in Triple Negative Breast Tumor Cells. Biomedicines 2023; 11:3066. [PMID: 38002066 PMCID: PMC10669784 DOI: 10.3390/biomedicines11113066] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Despite significant advances in the treatment of triple-negative breast cancer, this disease continues to pose a clinical challenge, with many patients ultimately suffering from relapse. Tumor cells that recover after entering into a state of senescence after chemotherapy or radiation have been shown to develop a more aggressive phenotype, and to contribute to disease recurrence. By combining the PARP inhibitor (PARPi), talazoparib, with radiation, senescence was enhanced in 4T1 and MDA-MB-231 triple-negative breast cancer cell lines (based on SA-β-gal upregulation, increased expression of CDKN1A and the senescence-associated secretory phenotype (SASP) marker, IL6). Subsequent treatment of the radiation- and talazoparib-induced senescent 4T1 and MDA-MB231 cells with navitoclax (ABT-263) resulted in significant apoptotic cell death. In immunocompetent tumor-bearing mice, navitoclax exerted a modest growth inhibitory effect when used alone, but dramatically interfered with the recovery of 4T1-derived tumors induced into senescence with ionizing radiation and talazoparib. These findings support the potential utility of a senolytic strategy in combination with the radiotherapy/PARPi combination to mitigate the risk of disease recurrence in triple-negative breast cancer.
Collapse
Affiliation(s)
- Abrar Softah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.); (M.R.A.); (A.R.A.); (K.A.); (M.M.A.); (S.A.); (N.A.A.)
| | - Moureq R. Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.); (M.R.A.); (A.R.A.); (K.A.); (M.M.A.); (S.A.); (N.A.A.)
| | - Ali R. Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.); (M.R.A.); (A.R.A.); (K.A.); (M.M.A.); (S.A.); (N.A.A.)
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan;
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.); (M.R.A.); (A.R.A.); (K.A.); (M.M.A.); (S.A.); (N.A.A.)
| | - Mashal M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.); (M.R.A.); (A.R.A.); (K.A.); (M.M.A.); (S.A.); (N.A.A.)
| | - Raed AlRowis
- Department of Periodontics and Community Dentistry, College of Dentistry, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Samiyah Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.); (M.R.A.); (A.R.A.); (K.A.); (M.M.A.); (S.A.); (N.A.A.)
| | - Norah A. Albekairy
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.); (M.R.A.); (A.R.A.); (K.A.); (M.M.A.); (S.A.); (N.A.A.)
| | - Hisashi Harada
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA;
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Rowan Boyd
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.B.); (E.C.)
| | - Eesha Chakraborty
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.B.); (E.C.)
| | - David A. Gewirtz
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.B.); (E.C.)
| | - Homood M. As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.); (M.R.A.); (A.R.A.); (K.A.); (M.M.A.); (S.A.); (N.A.A.)
| |
Collapse
|
10
|
Saleh T, Bloukh S, Hasan M, Al Shboul S. Therapy-induced senescence as a component of tumor biology: Evidence from clinical cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188994. [PMID: 37806641 DOI: 10.1016/j.bbcan.2023.188994] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/10/2023]
Abstract
Therapy-Induced Senescence (TIS) is an established response to anticancer therapy in a variety of cancer models. Ample evidence has characterized the triggers, hallmarks, and functional outcomes of TIS in preclinical studies; however, limited evidence delineates TIS in clinical cancer (human tumor samples). We examined the literature that investigated the induction of TIS in samples derived from human cancers and highlighted the major findings that suggested that TIS represents a main constituent of tumor biology. The most frequently utilized approach to identify TIS in human cancers was to investigate the protein expression of senescence-associated markers (such as cyclins, cyclin-dependent kinase inhibitors, Ki67, DNA damage repair response markers, DEC1, and DcR1) via immunohistochemical techniques using formalin-fixed paraffin-embedded (FFPE) tissue samples and/or testing the upregulation of Senescence-Associated β-galactosidase (SA-β-gal) in frozen sections of unfixed tumor samples. Collectively, and in studies where the extent of TIS was determined, TIS was detected in 31-66% of tumors exposed to various forms of chemotherapy. Moreover, TIS was not only limited to both malignant and non-malignant components of tumoral tissue but was also identified in samples of normal (non-transformed) tissue upon chemo- or radiotherapy exposure. Nevertheless, the available evidence continues to be limited and requires a more rigorous assessment of in vivo senescence based on novel approaches and more reliable molecular signatures. The accurate assessment of TIS will be beneficial for determining its relevant contribution to the overall outcome of cancer therapy and the potential translatability of senotherapeutics.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13115, Jordan.
| | - Sarah Bloukh
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Mira Hasan
- Department of Medicine, University of Connecticut Health Center, Farmington, USA
| | - Sofian Al Shboul
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13115, Jordan
| |
Collapse
|
11
|
Sun C, Chu A, Song R, Liu S, Chai T, Wang X, Liu Z. PARP inhibitors combined with radiotherapy: are we ready? Front Pharmacol 2023; 14:1234973. [PMID: 37954854 PMCID: PMC10637512 DOI: 10.3389/fphar.2023.1234973] [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: 06/05/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
PARP was an enzyme found in the nucleus of eukaryotic cells that played a crucial role in repairing damaged DNA. Recently, PARP inhibitors have demonstrated great potential in cancer treatment. Thus, the FDA has approved several small-molecule PARP inhibitors for cancer maintenance therapy. The combination of PARP inhibitors and radiotherapy relies on synthetic lethality, taking advantage of the flaws in DNA repair pathways to target cancer cells specifically. Studies conducted prior to clinical trials have suggested that the combination of PARP inhibitors and radiotherapy can enhance the sensitivity of cancer cells to radiation, intensify DNA damage, and trigger cell death. Combining radiotherapy with PARP inhibitors in clinical trials has enhanced the response rate and progression-free survival of diverse cancer patients. The theoretical foundation of PARP inhibitors combined with radiotherapy is explained in detail in this article, and the latest advances in preclinical and clinical research on these inhibitors for tumor radiotherapy are summarized. The problems in the current field are recognized in our research and potential therapeutic applications for tumors are suggested. Nevertheless, certain obstacles need to be tackled when implementing PARP inhibitors and radiotherapies in clinical settings. Factors to consider when using the combination therapy are the most suitable schedule and amount of medication, identifying advantageous candidates, and the probable adverse effects linked with the combination. The combination of radiotherapy and PARP inhibitors can greatly enhance the effectiveness of cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Wang
- Department of Radiation Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zongwen Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Al Shboul S, El-Sadoni M, Alhesa A, Abu Shahin N, Abuquteish D, Abu Al Karsaneh O, Alsharaiah E, Ismail MA, Tyutyunyk-Massey L, Alotaibi MR, Neely V, Harada H, Saleh T. NOXA expression is downregulated in human breast cancer undergoing incomplete pathological response and senescence after neoadjuvant chemotherapy. Sci Rep 2023; 13:15903. [PMID: 37741850 PMCID: PMC10517932 DOI: 10.1038/s41598-023-42994-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) is a frequently utilized approach to treat locally advanced breast cancer, but, unfortunately, a subset of tumors fails to undergo complete pathological response. Apoptosis and therapy-induced senescence (TIS) are both cell stress mechanisms but their exact role in mediating the pathological response to NAC is not fully elucidated. We investigated the change in expression of PAMIP1, the gene encoding for the pro-apoptotic protein, NOXA, following NAC in two breast cancer gene datasets, and the change in NOXA protein expression in response to NAC in 55 matched patient samples (pre- and post-NAC). PAMIP1 expression significantly declined in post-NAC in the two sets, and in our cohort, 75% of the samples exhibited a downregulation in NOXA post-NAC. Matched samples that showed a decline in NOXA post-NAC were examined for TIS based on a signature of downregulated expression of Lamin-B1 and Ki-67 and increased p16INK4a, and the majority exhibited a decrease in Lamin B1 (66%) and Ki-67 (80%), and increased p16INK4a (49%). Since our cohort consisted of patients that did not develop complete pathological response, such findings have clinical implications on the role of TIS and NOXA downregulation in mediating suboptimal responses to the currently established NAC.
Collapse
Affiliation(s)
- Sofian Al Shboul
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Mohammed El-Sadoni
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Ahmad Alhesa
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Nisreen Abu Shahin
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Dua Abuquteish
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Ola Abu Al Karsaneh
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Elham Alsharaiah
- Department of Pathology, King Hussein Medical Center, Royal Medical Service, Amman, 11942, Jordan
| | | | | | - Moureq R Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Victoria Neely
- Philips Institute for Oral Health Research, School of Dentistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Hisashi Harada
- Philips Institute for Oral Health Research, School of Dentistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan.
| |
Collapse
|
13
|
Gao Z, Zhao Q, Xu Y, Wang L. Improving the efficacy of combined radiotherapy and immunotherapy: focusing on the effects of radiosensitivity. Radiat Oncol 2023; 18:89. [PMID: 37226275 DOI: 10.1186/s13014-023-02278-5] [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] [Received: 04/09/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
Cancer treatment is gradually entering an era of precision, with multitude studies in gene testing and immunotherapy. Tumor cells can be recognized and eliminated by the immune system through the expression of tumor-associated antigens, but when the cancer escapes or otherwise suppresses immunity, the balance between cancer cell proliferation and immune-induced cancer cell killing may be interrupted, resulting in tumor proliferation and progression. There has been significant attention to combining conventional cancer therapies (i.e., radiotherapy) with immunotherapy as opposed to treatment alone. The combination of radio-immunotherapy has been demonstrated in both basic research and clinical trials to provide more effective anti-tumor responses. However, the absolute benefits of radio-immunotherapy are dependent on individual characteristics and not all patients can benefit from radio-immunotherapy. At present, there are numerous articles about exploring the optimal models for combination radio-immunotherapy, but the factors affecting the efficacy of the combination, especially with regard to radiosensitivity remain inconclusive. Radiosensitivity is a measure of the response of cells, tissues, or individuals to ionizing radiation, and various studies have shown that the radiosensitivity index (RSI) will be a potential biomarker for predicting the efficacy of combination radio-immunotherapy. The purpose of this review is to focus on the factors that influence and predict the radiosensitivity of tumor cells, and to evaluate the impact and predictive significance of radiosensitivity on the efficacy of radio-immunotherapy combination.
Collapse
Affiliation(s)
- Zhiru Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Qian Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430064, China
| | - Yiyue Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
14
|
Shi J, Sun Z, Gao Z, Huang D, Hong H, Gu J. Radioimmunotherapy in colorectal cancer treatment: present and future. Front Immunol 2023; 14:1105180. [PMID: 37234164 PMCID: PMC10206275 DOI: 10.3389/fimmu.2023.1105180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Colorectal cancer (CRC) is a deadly form of cancer worldwide. Patients with locally advanced rectal cancer and metastatic CRC have a poor long-term prognosis, and rational and effective treatment remains a major challenge. Common treatments include multi-modal combinations of surgery, radiotherapy, and chemotherapy; however, recurrence and metastasis rates remain high. The combination of radiotherapy and immunotherapy (radioimmunotherapy [RIT]) may offer new solutions to this problem, but its prospects remain uncertain. This review aimed to summarize the current applications of radiotherapy and immunotherapy, elaborate on the underlying mechanisms, and systematically review the preliminary results of RIT-related clinical trials for CRC. Studies have identified several key predictors of RIT efficacy. Summarily, rational RIT regimens can improve the outcomes of some patients with CRC, but current study designs have limitations. Further studies on RIT should focus on including larger sample sizes and optimizing the combination therapy regimen based on underlying influencing factors.
Collapse
Affiliation(s)
- Jingyi Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhuang Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhaoya Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Dandan Huang
- Department of Oncology, Peking University Shougang Hospital, Beijing, China
| | - Haopeng Hong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jin Gu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
- Peking Tsinghua Center for Life Science, Peking University International Cancer Center, Beijing, China
| |
Collapse
|
15
|
DeLuca VJ, Saleh T. Insights into the role of senescence in tumor dormancy: mechanisms and applications. Cancer Metastasis Rev 2023; 42:19-35. [PMID: 36681750 DOI: 10.1007/s10555-023-10082-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/12/2023] [Indexed: 01/23/2023]
Abstract
One of the most formidable challenges in oncology and tumor biology research is to provide an accurate understanding of tumor dormancy mechanisms. Dormancy refers to the ability of tumor cells to go undetected in the body for a prolonged period, followed by "spontaneous" escape. Various models of dormancy have been postulated, including angiogenic, immune-mediated, and cellular dormancy. While the former two propose mechanisms by which tumor growth may remain static at a population level, cellular dormancy refers to molecular processes that restrict proliferation at the cell level. Senescence is a form of growth arrest, during which cells undergo distinct phenotypic, epigenetic, and metabolic changes. Senescence is also associated with the development of a robust secretome, comprised of various chemokines and cytokines that interact with the surrounding microenvironment, including other tumor cells, stromal cells, endothelial cells, and immune cells. Both tumor and non-tumor cells can undergo senescence following various stressors, many of which are present during tumorigenesis and therapy. As such, senescent cells are present within forming tumors and in residual tumors post-treatment and therefore play a major role in tumor biology. However, the contributions of senescence to dormancy are largely understudied. Here, we provide an overview of multiple processes that have been well established as being involved in tumor dormancy, and we speculate on how senescence may contribute to these mechanisms.
Collapse
Affiliation(s)
- Valerie J DeLuca
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan.
| |
Collapse
|
16
|
Molecular targets that sensitize cancer to radiation killing: From the bench to the bedside. Biomed Pharmacother 2023; 158:114126. [PMID: 36521246 DOI: 10.1016/j.biopha.2022.114126] [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: 10/19/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Radiotherapy is a standard cytotoxic therapy against solid cancers. It uses ionizing radiation to kill tumor cells through damage to DNA, either directly or indirectly. Radioresistance is often associated with dysregulated DNA damage repair processes. Most radiosensitizers enhance radiation-mediated DNA damage and reduce the rate of DNA repair ultimately leading to accumulation of DNA damages, cell-cycle arrest, and cell death. Recently, agents targeting key signals in DNA damage response such as DNA repair pathways and cell-cycle have been developed. This new class of molecularly targeted radiosensitizing agents is being evaluated in preclinical and clinical studies to monitor their activity in potentiating radiation cytotoxicity of tumors and reducing normal tissue toxicity. The molecular pathways of DNA damage response are reviewed with a focus on the repair mechanisms, therapeutic targets under current clinical evaluation including ATM, ATR, CDK1, CDK4/6, CHK1, DNA-PKcs, PARP-1, Wee1, & MPS1/TTK and potential new targets (BUB1, and DNA LIG4) for radiation sensitization.
Collapse
|
17
|
Feng D, Shi X, You J, Xiong Q, Zhu W, Wei Q, Yang L. A cellular senescence-related gene prognostic index for biochemical recurrence and drug resistance in patients with prostate cancer. Am J Cancer Res 2022; 12:3811-3828. [PMID: 36119834 PMCID: PMC9441995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/12/2022] [Indexed: 06/15/2023] Open
Abstract
In this study, we aimed to establish a novel cellular senescence-related gene prognostic index (CSG PI) to predict biochemical recurrence (BCR) and drug resistance in patients with prostate cancer (PCa) undergoing radical radiotherapy or prostatectomy. We performed all analyses using R version 3.6.3 and its suitable packages. Cytoscape 3.8.2 was used to establish a network of transcription factors and competing endogenous RNAs. Three cellular senescence-related genes were used to establish the CSGPI. We observed that CSGPI was an independent risk factor for BCR in PCa patients (HR: 2.62; 95% CI: 1.55-4.44), consistent with the results of external validation (HR: 1.88; 95% CI: 1.12-3.14). The CSGPI had a moderate diagnostic effect on drug resistance (AUC: 0.812, 95% CI: 0.586-1.000). The lncRNA PART1 was significantly associated with BCR (HR: 0.46; 95% CI: 0.27-0.77), and might modulate the mRNA expression of definitive genes through interactions with 57 miRNAs. Gene set enrichment analysis indicated that CSGPI was closely related to ECM receptor interaction, focal adhesion, TGF beta signaling pathway, pathway in cancer, regulation of actin cytoskeleton, and so on. Immune checkpoint analysis showed that PDCD1LG2 and CD96 were significantly higher in the BCR group compared to non-BCR group, and patients with higher expression of CD96 were more prone to BCR than their counterparts (HR: 1.79; 95% CI: 1.06-3.03). In addition, the CSGPI score was significantly associated with the mRNA expression of HAVCR2, CD96, and CD47. Analysis of mismatch repair and methyltransferase genes showed that DNMT3B was more highly expressed in the BCR group and that patients with higher expression of DNMT3B experienced a higher risk of BCR (HR: 2.08; 95% CI: 1.23-3.52). We observed that M1 macrophage, CD8+ T cells, stromal score, immune score, and ESTIMATE score were higher in the BCR group. In contrast, tumor purity was less scored in the BCR group. Spearman analysis revealed a positive relationship between CSGPI and M1 macrophages, CD4+ T cells, dendritic cells, stromal score, immune score, and ESTIMATE score. In conclusion, we found that the CSGPI might serve as a biomarker to predict BCR and drug resistance in PCa patients. Moreover, CD96 and DNMT3B might be potential treatment targets, and immune evasion might contribute to the BCR process of PCa.
Collapse
Affiliation(s)
- Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University Chengdu 610041, Sichuan, People's Republic of China
| | - Xu Shi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University Chengdu 610041, Sichuan, People's Republic of China
| | - Jia You
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University Chengdu 610041, Sichuan, People's Republic of China
| | - Qiao Xiong
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University Chengdu 610041, Sichuan, People's Republic of China
| | - Weizhen Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University Chengdu 610041, Sichuan, People's Republic of China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University Chengdu 610041, Sichuan, People's Republic of China
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University Chengdu 610041, Sichuan, People's Republic of China
| |
Collapse
|
18
|
Effect of Autophagy Inhibitors on Radiosensitivity in DNA Repair-Proficient and -Deficient Glioma Cells. Medicina (B Aires) 2022; 58:medicina58070889. [PMID: 35888608 PMCID: PMC9317283 DOI: 10.3390/medicina58070889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/18/2023] Open
Abstract
Background and Objectives: The development of radioresistance is a fundamental barrier to successful glioblastoma therapy. Autophagy is thought to play a role in facilitating the DNA repair of DNA damage foci in radiation-exposed tumor cells, thus, potentially contributing to their restoration of proliferative capacity and development of resistance in vitro. However, the effect of autophagy inhibitors on DNA damage repair is not fully clear and requires further investigation. Materials and Methods: In this work, we utilized M059K (DNA-PKcs proficient) and M059J (DNA-PKcs deficient) glioma cell lines to investigate the role of autophagy inhibitors in the DNA repair of radiation-induced DNA damage. Cell viability following radiation was determined by trypan blue exclusion in both cell lines. Cell death and autophagy assays were performed to evaluate radiation-induced cell stress responses. DNA damage was measured as based on the intensity of phosphorylated γ-H2AX, a DNA double-stranded breaks (DSBs) marker, in the presence or absence of autophagy inhibitors. Results: The cell viability assay showed that M059J cells were more sensitive to the same dose of radiation (4 Gy) than M059K cells. This observation was accompanied by an elevation in γ-H2AX formation in M059J but not in M059K cells. In addition, the DAPI/TUNEL and Senescence-associated β-galactosidase (SA-β-gal) staining assays did not reveal significant differences in apoptosis and/or senescence induction in response to radiation, respectively, in either cell line. However, acridine orange staining demonstrated clear promotion of acidic vesicular organelles (AVOs) in both cell lines in response to 4 Gy radiation. Moreover, DNA damage marker levels were found to be elevated 72 h post-radiation when autophagy was inhibited by the lysosomotropic agent bafilomycin A1 (BafA1) or the PI3K inhibitor 3-methyl adenine (3-MA) in M059K cells. Conclusions: The extent of the DNA damage response remained high in the DNA-PKcs deficient cells following exposure to radiation, indicating their inability to repair the newly formed DNA-DSBs. On the other hand, radioresistant M059K cells showed more DNA damage response only when autophagy inhibitors were used with radiation, suggesting that the combination of autophagy inhibitors with radiation may interfere with DNA repair efficiency.
Collapse
|
19
|
Qin C, Ji Z, Zhai E, Xu K, Zhang Y, Li Q, Jing H, Wang X, Song X. PARP inhibitor olaparib enhances the efficacy of radiotherapy on XRCC2-deficient colorectal cancer cells. Cell Death Dis 2022; 13:505. [PMID: 35643812 PMCID: PMC9148313 DOI: 10.1038/s41419-022-04967-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/15/2022] [Accepted: 05/23/2022] [Indexed: 12/14/2022]
Abstract
The use of PARP inhibitors in combination with radiotherapy is a promising strategy to locally enhance DNA damage in tumors. Loss of XRCC2 compromises DNA damage repairs, and induced DNA damage burdens may increase the reliance on PARP-dependent DNA repairs of cancer cells to render cell susceptibility to PARP inhibitor therapy. Here we tested the hypothesis that XRCC2 loss sensitizes colorectal cancer (CRC) to PARP inhibitor in combination with radiotherapy (RT). We show that high levels of XRCC2 or PARP1 in LARC patients were significantly associated with poor overall survival (OS). Co-expression analyses found that low levels of PARP1 and XRCC2 were associated with better OS. Our in vitro experiments indicated that olaparib+IR led to reduced clonogenic survival, more DNA damage, and longer durations of cell cycle arrest and senescence in XRCC2-deficient cells relative to wild-type cells. Furthermore, our mouse xenograft experiments indicated that RT + olaparib had greater anti-tumor effects and led to long-term remission in mice with XRCC2-deficient tumors. These findings suggest that XRCC2-deficient CRC acquires high sensitivity to PARP inhibition after IR treatment and supports the clinical development for the use of olaparib as a radiosensitizer for treatment of XRCC2-deficient CRC.
Collapse
Affiliation(s)
- Changjiang Qin
- grid.256922.80000 0000 9139 560XDepartment of Gastrointestinal Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Zhiyu Ji
- grid.256922.80000 0000 9139 560XDepartment of Gastrointestinal Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Ertao Zhai
- grid.412615.50000 0004 1803 6239Department of Gastrointestinal and Pancreatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kaiwu Xu
- grid.412615.50000 0004 1803 6239Department of Gastrointestinal and Pancreatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yijie Zhang
- Department of Medical Oncology, Huaihe Hospital of Hennan University, Kaifeng, China
| | - Quanying Li
- grid.256922.80000 0000 9139 560XDepartment of Gastrointestinal Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Hong Jing
- Department of Pathology, Huaihe Hospital of Hennan University, Kaifeng, China
| | - Xiaoliang Wang
- grid.413087.90000 0004 1755 3939Department of General Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Xinming Song
- grid.412615.50000 0004 1803 6239Department of Gastrointestinal and Pancreatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
20
|
Lee JC, Kim GC, Lee NK, Kim SW, Cho YS, Chung SW, Lee YS, Chang HW, Byun Y, Kim SY. Feedback amplification of senolysis using caspase-3-cleavable peptide-doxorubicin conjugate and 2DG. J Control Release 2022; 346:158-168. [PMID: 35452763 DOI: 10.1016/j.jconrel.2022.04.012] [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] [Received: 07/27/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 11/29/2022]
Abstract
Therapy-induced senescence (TIS), a common outcome of current cancer therapy, is a known cause of late recurrence and metastasis and thus its eradication is crucial for therapy success. In this study, we introduced a conceptually novel strategy combining radiation-induced apoptosis-targeted chemotherapy (RIATC) with an effective glycolysis inhibitor, 2-deoxy-d-glucose (2DG) to target TIS. RIATC releases cytotoxic payload by amplification, continually increasing TIS, and this can be targeted by 2DG that stimulates an intrinsic apoptotic pathway in senescent cells, the senolysis; the senolytic 2DG also sensitizes cancer cells to chemo/radiation treatment. Anti-tumor efficacy of RIATC was investigated in numerous tumor models, and various cancer types were screened for TIS. Furthermore, in vitro evaluations of molecular markers of senescence, such as senescence-associated β-galactosidase (SA-β-Gal) assay, were performed to confirm that TIS was induced by RIATC therapy in MCF-7 cells. The combination therapy with 2DG proved to be effective in MCF-7 tumor-bearing mice that demonstrated feedback amplification of senolysis and successful inhibition of tumor growth. Our findings suggest that RIATC, when given together with 2DG, can overcome therapy-induced senescence and this combination is a promising strategy that enhances the therapeutic benefit of anti-cancer cytotoxic therapy.
Collapse
Affiliation(s)
- Jong Cheol Lee
- Department of Otorhinolaryngology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung-si, Gangwon-do 25440, Republic of Korea
| | - Gui Chul Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Na Kyeong Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Young Seok Cho
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung Woo Chung
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Yoon Se Lee
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Hyo Won Chang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea.
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| |
Collapse
|
21
|
Huart C, Fransolet M, Demazy C, Le Calvé B, Lucas S, Michiels C, Wéra AC. Taking Advantage of the Senescence-Promoting Effect of Olaparib after X-ray and Proton Irradiation Using the Senolytic Drug, ABT-263. Cancers (Basel) 2022; 14:cancers14061460. [PMID: 35326611 PMCID: PMC8946554 DOI: 10.3390/cancers14061460] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
Radiotherapy (RT) is a key component of cancer treatment. Although improvements have been made over the years, radioresistance remains a challenge. For this reason, a better understanding of cell fates in response to RT could improve therapeutic options to enhance cell death and reduce adverse effects. Here, we showed that combining RT (photons and protons) to noncytotoxic concentration of PARP inhibitor, Olaparib, induced a cell line-dependent senescence-like phenotype. The senescent cells were characterized by morphological changes, an increase in p21 mRNA expression as well as an increase in senescence-associated β-galactosidase activity. We demonstrated that these senescent cells could be specifically targeted by Navitoclax (ABT-263), a Bcl-2 family inhibitor. This senolytic drug led to significant cell death when combined with RT and Olaparib, while limited cytotoxicity was observed when used alone. These results demonstrate that a combination of RT with PARP inhibition and senolytics could be a promising therapeutic approach for cancer patients.
Collapse
Affiliation(s)
- Camille Huart
- Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (C.H.); (M.F.); (C.D.); (B.L.C.); (C.M.)
| | - Maude Fransolet
- Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (C.H.); (M.F.); (C.D.); (B.L.C.); (C.M.)
| | - Catherine Demazy
- Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (C.H.); (M.F.); (C.D.); (B.L.C.); (C.M.)
| | - Benjamin Le Calvé
- Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (C.H.); (M.F.); (C.D.); (B.L.C.); (C.M.)
| | - Stéphane Lucas
- Laboratory of Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium;
| | - Carine Michiels
- Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (C.H.); (M.F.); (C.D.); (B.L.C.); (C.M.)
| | - Anne-Catherine Wéra
- Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (C.H.); (M.F.); (C.D.); (B.L.C.); (C.M.)
- Molecular Imaging, Radiation and Oncology (MIRO) Lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Woluwe-Saint-Lambert, Belgium
- Correspondence:
| |
Collapse
|
22
|
Estrogen receptor inhibition mediates radiosensitization of ER-positive breast cancer models. NPJ Breast Cancer 2022; 8:31. [PMID: 35273179 PMCID: PMC8913671 DOI: 10.1038/s41523-022-00397-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022] Open
Abstract
Endocrine therapy (ET) is an effective first-line therapy for women with estrogen receptor-positive (ER + ) breast cancers. While both ionizing radiation (RT) and ET are used for the treatment of women with ER+ breast cancer, the most effective sequencing of therapy and the effect of ET on tumor radiosensitization remains unclear. Here we sought to understand the effects of inhibiting estrogen receptor (ER) signaling in combination with RT in multiple preclinical ER+ breast cancer models. Clonogenic survival assays were performed using variable pre- and post-treatment conditions to assess radiosensitization with estradiol, estrogen deprivation, tamoxifen, fulvestrant, or AZD9496 in ER+ breast cancer cell lines. Estrogen stimulation was radioprotective (radiation enhancement ratios [rER]: 0.51–0.82). Conversely, when given one hour prior to RT, ER inhibition or estrogen depletion radiosensitized ER+ MCF-7 and T47D cells (tamoxifen rER: 1.50–1.60, fulvestrant rER: 1.76–2.81, AZD9496 rER: 1.33–1.48, estrogen depletion rER: 1.47–1.51). Combination treatment resulted in an increase in double-strand DNA (dsDNA) breaks as a result of inhibition of non-homologous end joining-mediated dsDNA break repair with no effect on homologous recombination. Treatment with tamoxifen or fulvestrant in combination with RT also increased the number of senescent cells but did not affect apoptosis or cell cycle distribution. Using an MCF-7 xenograft model, concurrent treatment with tamoxifen and RT was synergistic and resulted in a significant decrease in tumor volume and a delay in time to tumor doubling without significant toxicity. These findings provide preclinical evidence that concurrent treatment with ET and RT may be an effective radiosensitization strategy.
Collapse
|
23
|
Ahmadinejad F, Bos T, Hu B, Britt E, Koblinski J, Souers AJ, Leverson JD, Faber AC, Gewirtz DA, Harada H. Senolytic-Mediated Elimination of Head and Neck Tumor Cells Induced Into Senescence by Cisplatin. Mol Pharmacol 2022; 101:168-180. [PMID: 34907000 PMCID: PMC8969145 DOI: 10.1124/molpharm.121.000354] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/25/2021] [Indexed: 01/10/2023] Open
Abstract
Therapeutic outcomes achieved in head and neck squamous cell carcinoma (HNSCC) patients by concurrent cisplatin-based chemoradiotherapy initially reflect both tumor regression and tumor stasis. However, local and distant metastasis and disease relapse are common in HNSCC patients. In the current work, we demonstrate that cisplatin treatment induces senescence in both p53 wild-type HN30 and p53 mutant HN12 head and neck cancer models. We also show that tumor cells can escape from senescence both in vitro and in vivo. We further establish the effectiveness of the senolytic, ABT-263 (Navitoclax), in elimination of senescent tumor cells after cisplatin treatment. Navitoclax increased apoptosis by 3.3-fold (P ≤ 0.05) at day 7 compared with monotherapy by cisplatin. Additionally, we show that ABT-263 interferes with the interaction between B-cell lymphoma-x large (BCL-XL) and BAX, anti- and pro-apoptotic proteins, respectively, followed by BAX activation, suggesting that ABT-263-induced apoptotic cell death is mediated through BAX. Our in vivo studies also confirm senescence induction in tumor cells by cisplatin, and the promotion of apoptosis coupled with a significant delay of tumor growth after sequential treatment with ABT-263. Sequential treatment with cisplatin followed by ABT-263 extended the humane endpoint to ∼130 days compared with cisplatin alone, where mice survived ∼75 days. These results support the premise that senolytic agents could be used to eliminate residual senescent tumor cells after chemotherapy and thereby potentially delay disease recurrence in head and neck cancer patients. SIGNIFICANCE STATEMENT: Disease recurrence is the most common cause of death in head and neck cancer patients. B-cell lymphoma-x large inhibitors such as ABT-263 (Navitoclax) have the capacity to be used in combination with cisplatin in head and neck cancer patients to eliminate senescent cells and possibly prevent disease relapse.
Collapse
Affiliation(s)
- Fereshteh Ahmadinejad
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - Tasia Bos
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - Bin Hu
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - Erin Britt
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - Jennifer Koblinski
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - Andrew J Souers
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - Joel D Leverson
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - Anthony C Faber
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - David A Gewirtz
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| | - Hisashi Harada
- Department of Human and Molecular Genetics, School of Medicine (F.A.), Philips Institute for Oral Health Research, School of Dentistry (T.B., E.B., A.C.F., H.H.), Cancer Mouse Models Core (B.H., J.K.), and Department of Pharmacology and Toxicology, School of Medicine (D.A.G.), Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; and AbbVie, North Chicago, Illinois (A.J.S., J.D.L.)
| |
Collapse
|
24
|
Russo M, Spagnuolo C, Moccia S, Tedesco I, Lauria F, Russo GL. Biochemical and Cellular Characterization of New Radio-Resistant Cell Lines Reveals a Role of Natural Flavonoids to Bypass Senescence. Int J Mol Sci 2021; 23:ijms23010301. [PMID: 35008725 PMCID: PMC8745286 DOI: 10.3390/ijms23010301] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/24/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the main causes of death worldwide, and, among the most frequent cancer types, osteosarcoma accounts for 56% of bone neoplasms observed in children and colorectal cancer for 10.2% of tumors diagnosed in the adult population. A common and frequent hurdle in cancer treatment is the emergence of resistance to chemo- and radiotherapy whose biological causes are largely unknown. In the present work, human osteosarcoma (SAOS) and colorectal adenocarcinoma (HT29) cell lines were γ-irradiated at doses mimicking the sub-lethal irradiation in clinical settings to obtain two radio-resistant cellular sub-populations named SAOS400 and HT500, respectively. Since “therapy-induced senescence” (TIS) is often associated with tumor response to radiotherapy in cancer cells, we measured specific cellular and biochemical markers of senescence in SAOS400 and HT500 cells. In detail, both cell lines were characterized by a higher level of expression of cyclin-dependent kinase inhibitors p16INK4 and p21CIP1 and increased positivity to SAβ-gal (senescence-associated β-galactosidase) with respect to parental cells. Moreover, the intracellular levels of reactive oxygen species in the resistant cells were significantly lower compared to the parental counterparts. Subsequently, we demonstrated that senolytic agents were able to sensitize SAOS400 and HT500 to cell death induced by γ-irradiation. Employing two natural flavonoids, fisetin and quercetin, and a BH3-mimetic, ABT-263/navitoclax, we observed that their association with γ-irradiation significantly reduced the expression of p16INK4, p21CIP1 and synergistically (combination index < 1) increased cell death compared to radiation mono-alone treatments. The present results reinforce the potential role of senolytics as adjuvant agents in cancer therapy.
Collapse
Affiliation(s)
- Maria Russo
- Correspondence: (M.R.); (G.L.R.); Tel.: +39-0825-299-331 (M.R.)
| | | | | | | | | | | |
Collapse
|
25
|
As Sobeai HM, Alohaydib M, Alhoshani AR, Alhazzani K, Almutairi MM, Saleh T, Gewirtz DA, Alotiabi MR. Sorafenib, rapamycin, and venetoclax attenuate doxorubicin-induced senescence and promote apoptosis in HCT116 cells. Saudi Pharm J 2021; 30:91-101. [PMID: 35145348 PMCID: PMC8802130 DOI: 10.1016/j.jsps.2021.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023] Open
Abstract
Emerging evidence has shown that the therapy-induced senescent growth arrest in cancer cells is of durable nature whereby a subset of cells can reinstate proliferative capacity. Promising new drugs named senolytics selectively target senescent cells and commit them into apoptosis. Accordingly, senolytics have been proposed as adjuvant cancer treatment to cull senescent tumor cells, and thus, screening for agents that exhibit senolytic properties is highly warranted. Our study aimed to investigate three agents, sorafenib, rapamycin, and venetoclax for their senolytic potential in doxorubicin-induced senescence in HCT116 cells. HCT116 cells were treated with one of the three agents, sorafenib (5 µM), rapamycin (100 nM), or venetoclax (10 µM), in the absence or presence of doxorubicin (1 µM). Senescence was evaluated using microscopy-based and flow cytometry-based Senescence-associated-β-galactosidase staining (SA-β-gal), while apoptosis was assessed using annexin V-FITC/PI, and Muse caspase-3/-7 activity assays. We screened for potential genes through which the three drugs exerted senolytic-like action using the Human Cancer Pathway Finder PCR array. The three agents reduced doxorubicin-induced senescent cell subpopulations and significantly enhanced the apoptotic effect of doxorubicin compared with those treated only with doxorubicin. The senescence genes IGFBP5 and BMI1 and the apoptosis genes CASP7 and CASP9 emerged as candidate genes through which the three drugs exhibited senolytic-like properties. These results suggest that the attenuation of doxorubicin-induced senescence might have shifted HCT116 cells to apoptosis by exposure to the tested pharmacological agents. Our work argues for the use of senolytics to reduce senescence-mediated resistance in tumor cells and to enhance chemotherapy efficacy.
Collapse
Affiliation(s)
- Homood M. As Sobeai
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Munirah Alohaydib
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ali R. Alhoshani
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mashal M. Almutairi
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - David A. Gewirtz
- Departments of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Moureq R. Alotiabi
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Corresponding author.
| |
Collapse
|
26
|
In-vitro 3D modelling for charged particle therapy - Uncertainties and opportunities. Adv Drug Deliv Rev 2021; 179:114018. [PMID: 34688685 DOI: 10.1016/j.addr.2021.114018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/07/2021] [Accepted: 10/18/2021] [Indexed: 12/26/2022]
Abstract
Radiation therapy is a critical component of oncologic management, with more than half of all cancer patients requiring radiotherapy at some point during their disease course. Over the last decade, there has been increasing interest in charged particle therapy due to its advantageous physical and radiobiologic properties, with the therapeutic use of proton beam therapy (PBT) expanding worldwide. However, there remain large gaps in our knowledge of the radiobiologic mechanisms that underlie key aspects of PBT, such as variations in relative biologic effectiveness (RBE), radioresistance, DNA damage response and repair pathways, as well as immunologic effects. In addition, while the emerging technique of ultra-high dose rate or FLASH radiotherapy, with its potential to further reduce normal tissue toxicities, is an exciting development, in-depth study is needed into the postulated biochemical mechanisms that underpin the FLASH effect such as the oxygen depletion hypothesis as well as the relative contributions of immune responses and the tumor microenvironment. Further investigation is also required to ensure that the FLASH effect is not diminished or lost in PBT. Current methods to evaluate the biologic effects of charged particle therapy rely heavily on 2D cell culture systems and/or animal models. However, both of these methods have well-recognized limitations which limit translatability of findings from bench to bedside. The advent of novel three-dimensional in-vitro tumor models offers a more physiologically relevant and high throughput in-vitro system for the study of tumor development as well as novel therapeutic approaches such as PBT. Advances in 3D cell culture methods, together with knowledge of disease mechanism, biomarkers, and genomic data, can be used to design personalized 3D models that most closely recapitulate tumor microenvironmental factors promoting a particular disease phenotype, moving 3D models and PBT into the age of precision medicine.
Collapse
|
27
|
Banerjee P, Kotla S, Reddy Velatooru L, Abe RJ, Davis EA, Cooke JP, Schadler K, Deswal A, Herrmann J, Lin SH, Abe JI, Le NT. Senescence-Associated Secretory Phenotype as a Hinge Between Cardiovascular Diseases and Cancer. Front Cardiovasc Med 2021; 8:763930. [PMID: 34746270 PMCID: PMC8563837 DOI: 10.3389/fcvm.2021.763930] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Overlapping risks for cancer and cardiovascular diseases (CVD), the two leading causes of mortality worldwide, suggest a shared biology between these diseases. The role of senescence in the development of cancer and CVD has been established. However, its role as the intersection between these diseases remains unclear. Senescence was originally characterized by an irreversible cell cycle arrest after a high number of divisions, namely replicative senescence (RS). However, it is becoming clear that senescence can also be instigated by cellular stress, so-called stress-induced premature senescence (SIPS). Telomere shortening is a hallmark of RS. The contribution of telomere DNA damage and subsequent DNA damage response/repair to SIPS has also been suggested. Although cellular senescence can mediate cell cycle arrest, senescent cells can also remain metabolically active and secrete cytokines, chemokines, growth factors, and reactive oxygen species (ROS), so-called senescence-associated secretory phenotype (SASP). The involvement of SASP in both cancer and CVD has been established. In patients with cancer or CVD, SASP is induced by various stressors including cancer treatments, pro-inflammatory cytokines, and ROS. Therefore, SASP can be the intersection between cancer and CVD. Importantly, the conventional concept of senescence as the mediator of cell cycle arrest has been challenged, as it was recently reported that chemotherapy-induced senescence can reprogram senescent cancer cells to acquire “stemness” (SAS: senescence-associated stemness). SAS allows senescent cancer cells to escape cell cycle arrest with strongly enhanced clonogenic growth capacity. SAS supports senescent cells to promote both cancer and CVD, particularly in highly stressful conditions such as cancer treatments, myocardial infarction, and heart failure. As therapeutic advances have increased overlapping risk factors for cancer and CVD, to further understand their interaction may provide better prevention, earlier detection, and safer treatment. Thus, it is critical to study the mechanisms by which these senescence pathways (SAS/SASP) are induced and regulated in both cancer and CVD.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Loka Reddy Velatooru
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Rei J Abe
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Elizabeth A Davis
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - John P Cooke
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Keri Schadler
- Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joerg Herrmann
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
28
|
Carpenter V, Saleh T, Min Lee S, Murray G, Reed J, Souers A, Faber AC, Harada H, Gewirtz DA. Androgen-deprivation induced senescence in prostate cancer cells is permissive for the development of castration-resistance but susceptible to senolytic therapy. Biochem Pharmacol 2021; 193:114765. [PMID: 34536356 DOI: 10.1016/j.bcp.2021.114765] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/30/2021] [Accepted: 09/13/2021] [Indexed: 01/26/2023]
Abstract
Prostate cancer (PCa) is one of the leading causes of cancer-related deaths in men. Although androgen deprivation therapies (ADT) and antiandrogens confer increased survival rates, most patients eventually develop castration resistant disease (CRPC). Previous studies have shown that these treatments have limited cytotoxicity, and instead, promote tumor cell growth arrest. We show here that PCa cells grown in either charcoal-stripped serum or exposed to the antiandrogen, bicalutamide, undergo a senescent growth arrest marked by morphological changes, upregulated senescence-associated-β-galactosidase (SA-β-Gal), cathepsin D accumulation, and expression of the senescence-associated secretory phenotype (SASP). The senescent growth arrest is, however, transient, as cells can resume proliferation upon restoration of normo-androgenic conditions. Intriguingly, enrichment for senescent cells confirmed that ADT-induced senescent cells recover their proliferative capacity, even under prolonged androgen deprivation, and form androgen-independent outgrowths. Transplantation of the enriched senescent population into castrated, syngeneic mice confirmed that senescent cells escape the growth arrest and form castration-resistant tumors in vivo. Outgrowth from senescence was associated with increased expression of constitutively active androgen receptor splice variants, a common mechanism of resistance to ADT. Finally, the selective elimination of senescent PCa cells following ADT in vitro by the senolytic navitoclax (ABT-263) interfered with the development of androgen-independent outgrowth. Taken together, these data support the premise that ADT-induced senescence is a transient cell state from which CRPC populations can emerge, identifying senescence as a potential driver of disease progression. Furthermore, it is feasible that senolytic therapy to eliminate senescent PCa cells could delay disease recurrence and/or progression to androgen independence.
Collapse
Affiliation(s)
- Valerie Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - So Min Lee
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Graeme Murray
- Department of Physics, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason Reed
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Physics, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew Souers
- AbbVie, 1 North Waukegan Road, North Chicago, IL, USA
| | - Anthony C Faber
- Philips Institute for Oral Health Research, School of Dentistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Hisashi Harada
- Philips Institute for Oral Health Research, School of Dentistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
29
|
Clark CA, Yang ES. Harnessing DNA Repair Defects to Augment Immune-Based Therapies in Triple-Negative Breast Cancer. Front Oncol 2021; 11:703802. [PMID: 34631532 PMCID: PMC8497895 DOI: 10.3389/fonc.2021.703802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has poor prognosis with limited treatment options, with little therapeutic progress made during the past several decades. DNA damage response (DDR) associated therapies, including radiation and inhibitors of DDR, demonstrate potential efficacy against TNBC, especially under the guidance of genomic subtype-directed treatment. The tumor immune microenvironment also contributes greatly to TNBC malignancy and response to conventional and targeted therapies. Immunotherapy represents a developing trend in targeted therapies directed against TNBC and strategies combining immunotherapy and modulators of the DDR pathways are being pursued. There is increasing understanding of the potential interplay between DDR pathways and immune-associated signaling. As such, the question of how we treat TNBC regarding novel immuno-molecular strategies is continually evolving. In this review, we explore the current and upcoming treatment options of TNBC in the context of DNA repair mechanisms and immune-based therapies, with a focus on implications of recent genomic analyses and clinical trial findings.
Collapse
Affiliation(s)
- Curtis A. Clark
- Department of Radiation Oncology, University of Alabama at Birmingham (UAB) School of Medicine, Birmingham, AL, United States
| | - Eddy S. Yang
- Department of Radiation Oncology, University of Alabama at Birmingham (UAB) School of Medicine, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham (UAB) School of Medicine, Birmingham, AL, United States
- Hugh Kaul Precision Medicine Institute, University of Alabama at Birmingham (UAB) School of Medicine, Birmingham, AL, United States
| |
Collapse
|
30
|
Waissi W, Amé JC, Mura C, Noël G, Burckel H. Gemcitabine-Based Chemoradiotherapy Enhanced by a PARP Inhibitor in Pancreatic Cancer Cell Lines. Int J Mol Sci 2021; 22:6825. [PMID: 34201963 PMCID: PMC8269291 DOI: 10.3390/ijms22136825] [Citation(s) in RCA: 8] [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: 04/27/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 01/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease with a 5-year overall survival of 9% for all stages. Gemcitabine-based chemoradiotherapy for locally advanced pancreatic cancer is highly toxic. We conducted an in vitro study to determine whether poly(ADP-ribose) polymerase-1 inhibition radiosensitized gemcitabine-based chemotherapy. Human pancreatic cancer cell lines, MIA PaCa-2, AsPC-1, BxPC-3 and PANC-1 were treated with gemcitabine (10 nM) and/or olaparib (1 µM). Low-LET gamma single dose of 2, 5 and 10 Gy radiations were carried out. Clonogenic assay, PAR immunoblotting, cell cycle distribution, γH2Ax, necrotic and autophagic cell death quantifications were performed. Treatment with olaparib alone was not cytotoxic, but highly radiosensitized cell lines, particularly at high dose per fraction A non-cytotoxic concentration of gemcitabine radiosensitized cells, but less than olaparib. Interestingly, olaparib significantly enhanced gemcitabine-based radiosensitization in PDAC cell lines with synergistic effect in BxPC-3 cell line. All cell lines were radiosensitized by the combination of gemcitabine and olaparib, through an increase of unrepaired double-strand, a G2 phase block and cell death. Radiosensitization was increased with high dose of radiation. The combination of olaparib with gemcitabine-based chemoradiotherapy could lead to an enhancement of local control in vivo and an improvement in disease-free survival.
Collapse
Affiliation(s)
- Waisse Waissi
- Department of Radiation Oncology, Centre Leon Bérard, 69008 Lyon, France;
| | - Jean-Christophe Amé
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire d’Excellence Medalis, UMR7242, Centre Nationale de la Recherche Scientifique/Université de Strasbourg, Institut de Recherche de l’Ecole de Biotechnologie de Strasbourg, 300 bld. S. Brant, CS10413, 67412 Illkirch, France;
| | - Carole Mura
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg University, UNICANCER, Paul Strauss Comprehensive Cancer Center, Radiobiology Laboratory, 67000 Strasbourg, France;
| | - Georges Noël
- Institut de Cancérologie Strasbourg Europe (ICANS), UNICANCER, Paul Strauss Comprehensive Cancer Center, Department of Radiation Oncology, 17 Rue Albert Calmette, 67200 Strasbourg, France;
- Strasbourg University, CNRS, IPHC, UMR 7178, 67200 Strasbourg, France
| | - Hélène Burckel
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg University, UNICANCER, Paul Strauss Comprehensive Cancer Center, Radiobiology Laboratory, 67000 Strasbourg, France;
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW To summarize recently discovered PARP inhibitor resistance mechanisms and highlight the clinical relevance of these findings to date. RECENT FINDINGS A predominant mechanism of acquired PARP inhibitor resistance in homologous recombination-deficient cancers is the acquisition of homologous recombination proficiency as a consequence of secondary genetic or epigenetic events, such as secondary mutations in BRCA1 or BRCA2, or reversal of BRCA1 promoter methylation that restores homologous recombination and leads to PARP inhibitor resistance. Multiple other potential mechanisms of acquired resistance to PARP inhibitors including loss of DNA end resection inhibition (53BP1/REV7/RIF1/Sheldin) or DNA replication fork protection (PTIP/EZH2), but also increased drug efflux or induction of a reversible senescent or mesenchymal cell state have been described in ovarian cancer models. However, only few of these mechanisms have been identified in clinical samples. SUMMARY Multiple adaptive responses following PARP inhibitor treatment have been identified. Further research is needed to better understand what role these mechanisms play for clinical PARP inhibitor resistance and how these mechanisms may render ovarian cancer cells susceptible to subsequent novel combination therapies.
Collapse
|
32
|
Demény MA, Virág L. The PARP Enzyme Family and the Hallmarks of Cancer Part 1. Cell Intrinsic Hallmarks. Cancers (Basel) 2021; 13:cancers13092042. [PMID: 33922595 PMCID: PMC8122967 DOI: 10.3390/cancers13092042] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/02/2021] [Accepted: 04/20/2021] [Indexed: 12/21/2022] Open
Abstract
The 17-member poly (ADP-ribose) polymerase enzyme family, also known as the ADP-ribosyl transferase diphtheria toxin-like (ARTD) enzyme family, contains DNA damage-responsive and nonresponsive members. Only PARP1, 2, 5a, and 5b are capable of modifying their targets with poly ADP-ribose (PAR) polymers; the other PARP family members function as mono-ADP-ribosyl transferases. In the last decade, PARP1 has taken center stage in oncology treatments. New PARP inhibitors (PARPi) have been introduced for the targeted treatment of breast cancer 1 or 2 (BRCA1/2)-deficient ovarian and breast cancers, and this novel therapy represents the prototype of the synthetic lethality paradigm. Much less attention has been paid to other PARPs and their potential roles in cancer biology. In this review, we summarize the roles played by all PARP enzyme family members in six intrinsic hallmarks of cancer: uncontrolled proliferation, evasion of growth suppressors, cell death resistance, genome instability, reprogrammed energy metabolism, and escape from replicative senescence. In a companion paper, we will discuss the roles of PARP enzymes in cancer hallmarks related to cancer-host interactions, including angiogenesis, invasion and metastasis, evasion of the anticancer immune response, and tumor-promoting inflammation. While PARP1 is clearly involved in all ten cancer hallmarks, an increasing body of evidence supports the role of other PARPs in modifying these cancer hallmarks (e.g., PARP5a and 5b in replicative immortality and PARP2 in cancer metabolism). We also highlight controversies, open questions, and discuss prospects of recent developments related to the wide range of roles played by PARPs in cancer biology. Some of the summarized findings may explain resistance to PARPi therapy or highlight novel biological roles of PARPs that can be therapeutically exploited in novel anticancer treatment paradigms.
Collapse
Affiliation(s)
- Máté A. Demény
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: (M.A.D.); (L.V.)
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: (M.A.D.); (L.V.)
| |
Collapse
|
33
|
Saoudaoui S, Bernard M, Cardin GB, Malaquin N, Christopoulos A, Rodier F. mTOR as a senescence manipulation target: A forked road. Adv Cancer Res 2021; 150:335-363. [PMID: 33858600 DOI: 10.1016/bs.acr.2021.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cellular senescence, cancer and aging are highly interconnected. Among many important molecular machines that lie at the intersection of this triad, the mechanistic (formerly mammalian) target of rapamycin (mTOR) is a central regulator of cell metabolism, proliferation, and survival. The mTOR signaling cascade is essential to maintain cellular homeostasis in normal biological processes or in response to stress, and its dysregulation is implicated in the progression of many disorders, including age-associated diseases. Accordingly, the pharmacological implications of mTOR inhibition using rapamycin or others rapalogs span the treatment of various human diseases from immune disorders to cancer. Importantly, rapamycin is one of the only known pan-species drugs that can extend lifespan. The molecular and cellular mechanisms explaining the phenotypic consequences of mTOR are vast and heavily studied. In this review, we will focus on the potential role of mTOR in the context of cellular senescence, a tumor suppressor mechanism and a pillar of aging. We will explore the link between senescence, autophagy and mTOR and discuss the opportunities to exploit senescence-associated mTOR functions to manipulate senescence phenotypes in age-associated diseases and cancer treatment.
Collapse
Affiliation(s)
- Sarah Saoudaoui
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Monique Bernard
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Guillaume B Cardin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Apostolos Christopoulos
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada; Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada; Université de Montréal, Département de radiologie, radio-oncologie et médicine nucléaire, Montreal, QC, Canada.
| |
Collapse
|
34
|
Patel NH, Bloukh S, Alwohosh E, Alhesa A, Saleh T, Gewirtz DA. Autophagy and senescence in cancer therapy. Adv Cancer Res 2021; 150:1-74. [PMID: 33858594 DOI: 10.1016/bs.acr.2021.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tumor cells can undergo diverse responses to cancer therapy. While apoptosis represents the most desirable outcome, tumor cells can alternatively undergo autophagy and senescence. Both autophagy and senescence have the potential to make complex contributions to tumor cell survival via both cell autonomous and cell non-autonomous pathways. The induction of autophagy and senescence in tumor cells, preclinically and clinically, either individually or concomitantly, has generated interest in the utilization of autophagy modulating and senolytic therapies to target autophagy and senescence, respectively. This chapter summarizes the current evidence for the promotion of autophagy and senescence as fundamental responses to cancer therapy and discusses the complexity of their functional contributions to cell survival and disease outcomes. We also highlight current modalities designed to exploit autophagy and senescence in efforts to improve the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Nipa H Patel
- Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, VA, United States; Massey Cancer Center, Goodwin Research Laboratories, Virginia Commonwealth University, Richmond, VA, United States
| | - Sarah Bloukh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Enas Alwohosh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Ahmad Alhesa
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - David A Gewirtz
- Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, VA, United States; Massey Cancer Center, Goodwin Research Laboratories, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
35
|
Abstract
The use of DNA-damaging agents such as radiotherapy and chemotherapy has been a mainstay treatment protocol for many cancers, including lung and prostate. Recently, FDA approval of inhibitors of DNA repair, and targeting innate immunity to enhance the efficacy of DNA-damaging agents have gained much attention. Yet, inherent or acquired resistance against DNA-damaging therapies persists as a fundamental drawback. While cancer eradication by causing cancer cell death through induction of apoptosis is the ultimate goal of anti-cancer treatments, autophagy and senescence are two major cellular responses induced by clinically tolerable doses of DNA-damaging therapies. Unlike apoptosis, autophagy and senescence can act as both pro-tumorigenic as well as tumor suppressive mechanisms. DNA damage-induced senescence is associated with a pro-inflammatory secretory phenotype, which contributes to reshaping the tumor- immune microenvironment. Moreover, PTEN (phosphatase and tensin homolog) is a tumor supressor deleted in many tumors, and has been implicated in both senescence and autophagy. This review presents an overview of the literature on the regulation and consequences of DNA damage- induced senescence in cancer cells, with a specific focus on autophagy and PTEN. Both autophagy and senescence occur concurrently in the same cells in response to DNA damaging agents. However, a deterministic relationship between these fundamental processes has been controversial. We present experimental evidence obtained with tumor cells, with a prime focus on two models of cancer, prostate and lung. A better understanding of mechanisms associated with DNA damage-induced cellular senescence is central to fully exploit the potential of DNA-damaging agents against cancer.
Collapse
Affiliation(s)
- Arishya Sharma
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.
| | - Alexandru Almasan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States; Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.
| |
Collapse
|
36
|
Senolytics for Cancer Therapy: Is All That Glitters Really Gold? Cancers (Basel) 2021; 13:cancers13040723. [PMID: 33578753 PMCID: PMC7916462 DOI: 10.3390/cancers13040723] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Senescence is an essential component of tumor cell biology and is a primary cell stress response to therapy. While the long-term impact of senescence in cancer therapy is not yet fully understood, the use of senolytics, drugs that selectively kill senescent cells, is an area of active investigation in cancer treatment. Several challenges and unanswered questions have arisen from the current preclinical literature, indicating the need to re-evaluate some of the basic premises and experimental approaches, as well as the potential utility for translating to the clinic the application of senolytics as adjuvants to current cancer therapy. Abstract Senolytics represent a group of mechanistically diverse drugs that can eliminate senescent cells, both in tumors and in several aging-related pathologies. Consequently, senolytic use has been proposed as a potential adjuvant approach to improve the response to senescence-inducing conventional and targeted cancer therapies. Despite the unequivocal promise of senolytics, issues of universality, selectivity, resistance, and toxicity remain to be further clarified. In this review, we attempt to summarize and analyze the current preclinical literature involving the use of senolytics in senescent tumor cell models, and to propose tenable solutions and future directions to improve the understanding and use of this novel class of drugs.
Collapse
|
37
|
Bryson BL, Tamagno I, Taylor SE, Parameswaran N, Chernosky NM, Balasubramaniam N, Jackson MW. Aberrant Induction of a Mesenchymal/Stem Cell Program Engages Senescence in Normal Mammary Epithelial Cells. Mol Cancer Res 2020; 19:651-666. [PMID: 33443106 DOI: 10.1158/1541-7786.mcr-19-1181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 10/23/2020] [Accepted: 12/15/2020] [Indexed: 11/16/2022]
Abstract
Although frequently associated with tumor progression, inflammatory cytokines initially restrain transformation by inducing senescence, a key tumor-suppressive barrier. Here, we demonstrate that the inflammatory cytokine, oncostatin M, activates a mesenchymal/stem cell (SC) program that engages cytokine-induced senescence (CIS) in normal human epithelial cells. CIS is driven by Snail induction and requires cooperation between STAT3 and the TGFβ effector, SMAD3. Importantly, as cells escape CIS, they retain the mesenchymal/SC program and are thereby bestowed with a set of cancer SC (CSC) traits. Of therapeutic importance, cells that escape CIS can be induced back into senescence by CDK4/6 inhibition, confirming that the mechanisms allowing cells to escape senescence are targetable and reversible. Moreover, by combining CDK4/6 inhibition with a senolytic therapy, mesenchymal/CSCs can be efficiently killed. Our studies provide insight into how the CIS barriers that prevent tumorigenesis can be exploited as potential therapies for highly aggressive cancers. IMPLICATIONS: These studies reveal how a normal cell's arduous escape from senescence can bestow aggressive features early in the transformation process, and how this persistent mesenchymal/SC program can create a novel potential targetability following tumor development.
Collapse
Affiliation(s)
- Benjamin L Bryson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Sarah E Taylor
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Neetha Parameswaran
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Noah M Chernosky
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Nikhila Balasubramaniam
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Mark W Jackson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
38
|
Yusoh NA, Ahmad H, Gill MR. Combining PARP Inhibition with Platinum, Ruthenium or Gold Complexes for Cancer Therapy. ChemMedChem 2020; 15:2121-2135. [PMID: 32812709 PMCID: PMC7754470 DOI: 10.1002/cmdc.202000391] [Citation(s) in RCA: 25] [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: 06/04/2020] [Indexed: 12/24/2022]
Abstract
Platinum drugs are heavily used first-line chemotherapeutic agents for many solid tumours and have stimulated substantial interest in the biological activity of DNA-binding metal complexes. These complexes generate DNA lesions which trigger the activation of DNA damage response (DDR) pathways that are essential to maintain genomic integrity. Cancer cells exploit this intrinsic DNA repair network to counteract many types of chemotherapies. Now, advances in the molecular biology of cancer has paved the way for the combination of DDR inhibitors such as poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) and agents that induce high levels of DNA replication stress or single-strand break damage for synergistic cancer cell killing. In this review, we summarise early-stage, preclinical and clinical findings exploring platinum and emerging ruthenium anti-cancer complexes alongside PARPi in combination therapy for cancer and also describe emerging work on the ability of ruthenium and gold complexes to directly inhibit PARP activity.
Collapse
Affiliation(s)
- Nur Aininie Yusoh
- Department of ChemistryFaculty of ScienceUniversiti Putra Malaysia43400 UPMSerdang, SelangorMalaysia
| | - Haslina Ahmad
- Department of ChemistryFaculty of ScienceUniversiti Putra Malaysia43400 UPMSerdang, SelangorMalaysia
- Integrated Chemical BiophysicsFaculty of ScienceUniversiti Putra Malaysia43400 UPMSerdang, SelangorMalaysia
| | - Martin R. Gill
- Department of ChemistrySwansea UniversitySwanseaWales (UK
| |
Collapse
|
39
|
Saleh T, Carpenter VJ, Tyutyunyk‐Massey L, Murray G, Leverson JD, Souers AJ, Alotaibi MR, Faber AC, Reed J, Harada H, Gewirtz DA. Clearance of therapy-induced senescent tumor cells by the senolytic ABT-263 via interference with BCL-X L -BAX interaction. Mol Oncol 2020; 14:2504-2519. [PMID: 32652830 PMCID: PMC7530780 DOI: 10.1002/1878-0261.12761] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/11/2020] [Accepted: 07/07/2020] [Indexed: 01/14/2023] Open
Abstract
Tumor cells undergo senescence in response to both conventional and targeted cancer therapies. The induction of senescence in response to cancer therapy can contribute to unfavorable patient outcomes, potentially including disease relapse. This possibiliy is supported by our findings that tumor cells induced into senescence by doxorubicin or etoposide can give rise to viable tumors in vivo. We further demonstrate sensitivity of these senescent tumor cells to the senolytic ABT-263 (navitoclax), therefore providing a "two-hit" approach to eliminate senescent tumor cells that persist after exposure to chemotherapy or radiation. The sequential combination of therapy-induced senescence and ABT-263 could shift the response to therapy toward apoptosis by interfering with the interaction between BCL-XL and BAX. The administration of ABT-263 after either etoposide or doxorubicin also resulted in marked, prolonged tumor suppression in tumor-bearing animals. These findings support the premise that senolytic therapy following conventional cancer therapy may improve therapeutic outcomes and delay disease recurrence.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Basic Medical SciencesFaculty of MedicineThe Hashemite UniversityZarqaJordan
- Departments of Pharmacology & ToxicologySchool of MedicineMassey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - Valerie J. Carpenter
- Departments of Pharmacology & ToxicologySchool of MedicineMassey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - Liliya Tyutyunyk‐Massey
- Departments of Pharmacology & ToxicologySchool of MedicineMassey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - Graeme Murray
- Department of PhysicsVirginia Commonwealth UniversityRichmondVAUSA
| | | | | | - Moureq R. Alotaibi
- Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Anthony C. Faber
- Philips Institute for Oral Health ResearchSchool of DentistryMassey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - Jason Reed
- Department of PhysicsVirginia Commonwealth UniversityRichmondVAUSA
| | - Hisashi Harada
- Philips Institute for Oral Health ResearchSchool of DentistryMassey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - David A. Gewirtz
- Departments of Pharmacology & ToxicologySchool of MedicineMassey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
| |
Collapse
|
40
|
Carpenter VJ, Patel BB, Autorino R, Smith SC, Gewirtz DA, Saleh T. Senescence and castration resistance in prostate cancer: A review of experimental evidence and clinical implications. Biochim Biophys Acta Rev Cancer 2020; 1874:188424. [PMID: 32956765 DOI: 10.1016/j.bbcan.2020.188424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/10/2023]
Abstract
The development of Castration-Resistant Prostate Cancer (CRPC) remains a major challenge in the treatment of this disease. While Androgen Deprivation Therapy (ADT) can result in tumor shrinkage, a primary response of Prostate Cancer (PCa) cells to ADT is a senescent growth arrest. As a response to cancer therapies, senescence has often been considered as a beneficial outcome due to its association with stable growth abrogation, as well as the potential for immune system activation via the Senescence-Associated Secretory Phenotype (SASP). However, there is increasing evidence that not only can senescent cells regain proliferative capacity, but that senescence contributes to deleterious effects of cancer chemotherapy, including disease recurrence. Notably, the preponderance of work investigating the consequences of therapy-induced senescence on tumor progression has been performed in non-PCa models. Here, we summarize the evidence that ADT promotes a senescent response in PCa and postulate mechanisms by which senescence may contribute to the development of castration-resistance. Primarily, we suggest that ADT-induced senescence may support CRPC development via escape from senescence, by cell autonomous-reprogramming, and by the formation of a pro-tumorigenic SASP. However, due to the scarcity of direct evidence from PCa models, the consequences of ADT-induced senescence outlined here remain speculative until the relationship between senescence and CRPC can be experimentally defined.
Collapse
Affiliation(s)
- Valerie J Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Bhaumik B Patel
- Department of Internal Medicine, Division of Hematology, Oncology & Palliative Care, VCU Health, Richmond, VA, USA
| | - Riccardo Autorino
- Department of Surgery, Division of Urology, VCU Health, Richmond, VA, USA
| | | | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tareq Saleh
- The Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan.
| |
Collapse
|
41
|
Patel NH, Sohal SS, Manjili MH, Harrell JC, Gewirtz DA. The Roles of Autophagy and Senescence in the Tumor Cell Response to Radiation. Radiat Res 2020; 194:103-115. [PMID: 32845995 PMCID: PMC7482104 DOI: 10.1667/rade-20-00009] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/15/2020] [Indexed: 01/10/2023]
Abstract
Radiation is a critical pillar in cancer therapeutics, exerting its anti-tumor DNA-damaging effects through various direct and indirect mechanisms. Radiation has served as an effective mode of treatment for a number of cancer types, providing both curative and palliative treatment; however, resistance to therapy persists as a fundamental limitation. While cancer cell death is the ideal outcome of any anti-tumor treatment, radiation induces several responses, including apoptotic cell death, mitotic catastrophe, autophagy and senescence, where autophagy and senescence may promote cell survival. In most cases, autophagy, a conventionally cytoprotective mechanism, is a "first" responder to damage incurred from chemotherapy and radiation treatment. The paradigm developed on the premise that autophagy is cytoprotective in nature has provided the rationale for current clinical trials designed with the goal of radiosensitizing cancer cells through the use of autophagy inhibitors; however, these have failed to produce consistent results. Delving further into pre-clinical studies, autophagy has actually been shown to take diverse, sometimes opposing, forms, such as acting in a cytotoxic or nonprotective fashion, which may be partially responsible for the inconsistency of clinical outcomes. Furthermore, autophagy can have both pro- and anti-tumorigenic effects, while also having an important immune modulatory function. Senescence often occurs in tandem with autophagy, which is also the case with radiation. Radiation-induced senescence is frequently followed by a phase of proliferative recovery in a subset of cells and has been proposed as a tumor dormancy model, which can contribute to resistance to therapy and possibly also disease recurrence. Senescence induction is often accompanied by a unique secretory phenotype that can either promote or suppress immune functions, depending on the expression profile of cytokines and chemokines. Novel therapeutics selectively cytotoxic to senescent cells (senolytics) may prove to prolong remission by delaying disease recurrence in patients. Accurate assessment of primary responses to radiation may provide potential targets that can be manipulated for therapeutic benefit to sensitize cancer cells to radiotherapy, while sparing normal tissue.
Collapse
Affiliation(s)
- Nipa H. Patel
- Departments of Pharmacology and Toxicology, Richmond, Virginia 23298
| | - Sahib S. Sohal
- Departments of Pathology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Masoud H Manjili
- Departments of Microbiology and Immunology, Massey Cancer Center, Richmond, Virginia 23298
| | - J. Chuck Harrell
- Departments of Pathology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - David A. Gewirtz
- Departments of Pharmacology and Toxicology, Richmond, Virginia 23298
| |
Collapse
|
42
|
Regulation of DNA Damage Response and Homologous Recombination Repair by microRNA in Human Cells Exposed to Ionizing Radiation. Cancers (Basel) 2020; 12:cancers12071838. [PMID: 32650508 PMCID: PMC7408912 DOI: 10.3390/cancers12071838] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Ionizing radiation may be of both artificial and natural origin and causes cellular damage in living organisms. Radioactive isotopes have been used significantly in cancer therapy for many years. The formation of DNA double-strand breaks (DSBs) is the most dangerous effect of ionizing radiation on the cellular level. After irradiation, cells activate a DNA damage response, the molecular path that determines the fate of the cell. As an important element of this, homologous recombination repair is a crucial pathway for the error-free repair of DNA lesions. All components of DNA damage response are regulated by specific microRNAs. MicroRNAs are single-stranded short noncoding RNAs of 20–25 nt in length. They are directly involved in the regulation of gene expression by repressing translation or by cleaving target mRNA. In the present review, we analyze the biological mechanisms by which miRNAs regulate cell response to ionizing radiation-induced double-stranded breaks with an emphasis on DNA repair by homologous recombination, and its main component, the RAD51 recombinase. On the other hand, we discuss the ability of DNA damage response proteins to launch particular miRNA expression and modulate the course of this process. A full understanding of cell response processes to radiation-induced DNA damage will allow us to develop new and more effective methods of ionizing radiation therapy for cancers, and may help to develop methods for preventing the harmful effects of ionizing radiation on healthy organisms.
Collapse
|
43
|
Ghorai A, Mahaddalkar T, Thorat R, Dutt S. Sustained inhibition of PARP-1 activity delays glioblastoma recurrence by enhancing radiation-induced senescence. Cancer Lett 2020; 490:44-53. [PMID: 32645394 DOI: 10.1016/j.canlet.2020.06.023] [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: 10/10/2019] [Revised: 06/08/2020] [Accepted: 06/26/2020] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most common primary brain tumor and is highly aggressive with a median survival of 15 months. We have previously shown that residual cells of GBM form multinucleated giant cells (MNGCs) showing a senescent phenotype, but eventually escape from therapy induced senescence (TIS), resulting in GBM recurrence. Here we demonstrate the role of PARP-1 in TIS and its recovery. We show that genetic and pharmacological inhibition of PARP-1 has an anti-proliferative effect on GBM cell lines and primary cultures derived from patient samples. Furthermore, the PARP-1 inhibitor olaparib, in combination with radiation increased MNGCs formation and senescence as assessed by β-galactosidase activity, and macroH2A1 levels in residual cells. Additionally, we found that reduced PARP-1 activity and not protein levels in residual cells was crucial for MNGCs formation and their maintenance in the senescent state. PARP-1 activity was restored to higher levels in recurrent cells that escaped from TIS. Importantly, olaparib + radiation treatment significantly delayed recurrence in vitro as well in vivo in orthotopic GBM mouse models with a significant increase in overall survival of mice. Overall, this study demonstrates that sustained inhibition of PARP-1 activity during radiation treatment significantly delays GBM recurrence.
Collapse
Affiliation(s)
- Atanu Ghorai
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Tejashree Mahaddalkar
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India.
| |
Collapse
|
44
|
Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther 2020; 5:60. [PMID: 32355263 PMCID: PMC7192953 DOI: 10.1038/s41392-020-0150-x] [Citation(s) in RCA: 641] [Impact Index Per Article: 128.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/20/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy is one of the most common countermeasures for treating a wide range of tumors. However, the radioresistance of cancer cells is still a major limitation for radiotherapy applications. Efforts are continuously ongoing to explore sensitizing targets and develop radiosensitizers for improving the outcomes of radiotherapy. DNA double-strand breaks are the most lethal lesions induced by ionizing radiation and can trigger a series of cellular DNA damage responses (DDRs), including those helping cells recover from radiation injuries, such as the activation of DNA damage sensing and early transduction pathways, cell cycle arrest, and DNA repair. Obviously, these protective DDRs confer tumor radioresistance. Targeting DDR signaling pathways has become an attractive strategy for overcoming tumor radioresistance, and some important advances and breakthroughs have already been achieved in recent years. On the basis of comprehensively reviewing the DDR signal pathways, we provide an update on the novel and promising druggable targets emerging from DDR pathways that can be exploited for radiosensitization. We further discuss recent advances identified from preclinical studies, current clinical trials, and clinical application of chemical inhibitors targeting key DDR proteins, including DNA-PKcs (DNA-dependent protein kinase, catalytic subunit), ATM/ATR (ataxia-telangiectasia mutated and Rad3-related), the MRN (MRE11-RAD50-NBS1) complex, the PARP (poly[ADP-ribose] polymerase) family, MDC1, Wee1, LIG4 (ligase IV), CDK1, BRCA1 (BRCA1 C terminal), CHK1, and HIF-1 (hypoxia-inducible factor-1). Challenges for ionizing radiation-induced signal transduction and targeted therapy are also discussed based on recent achievements in the biological field of radiotherapy.
Collapse
Affiliation(s)
- Rui-Xue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, 410078, Changsha, People's Republic of China
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, 100850, Beijing, People's Republic of China.
- Institute for Chemical Carcinogenesis, State Key Laboratory of Respiratory, Guangzhou Medical University, 511436, Guangzhou, People's Republic of China.
| |
Collapse
|
45
|
Saleh T, Bloukh S, Carpenter VJ, Alwohoush E, Bakeer J, Darwish S, Azab B, Gewirtz DA. Therapy-Induced Senescence: An "Old" Friend Becomes the Enemy. Cancers (Basel) 2020; 12:cancers12040822. [PMID: 32235364 PMCID: PMC7226427 DOI: 10.3390/cancers12040822] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 01/10/2023] Open
Abstract
For the past two decades, cellular senescence has been recognized as a central component of the tumor cell response to chemotherapy and radiation. Traditionally, this form of senescence, termed Therapy-Induced Senescence (TIS), was linked to extensive nuclear damage precipitated by classical genotoxic chemotherapy. However, a number of other forms of therapy have also been shown to induce senescence in tumor cells independently of direct genomic damage. This review attempts to provide a comprehensive summary of both conventional and targeted anticancer therapeutics that have been shown to induce senescence in vitro and in vivo. Still, the utility of promoting senescence as a therapeutic endpoint remains under debate. Since senescence represents a durable form of growth arrest, it might be argued that senescence is a desirable outcome of cancer therapy. However, accumulating evidence suggesting that cells have the capacity to escape from TIS would support an alternative conclusion, that senescence provides an avenue whereby tumor cells can evade the potentially lethal action of anticancer drugs, allowing the cells to enter a temporary state of dormancy that eventually facilitates disease recurrence, often in a more aggressive state. Furthermore, TIS is now strongly connected to tumor cell remodeling, potentially to tumor dormancy, acquiring more ominous malignant phenotypes and accounts for several untoward adverse effects of cancer therapy. Here, we argue that senescence represents a barrier to effective anticancer treatment, and discuss the emerging efforts to identify and exploit agents with senolytic properties as a strategy for elimination of the persistent residual surviving tumor cell population, with the goal of mitigating the tumor-promoting influence of the senescent cells and to thereby reduce the likelihood of cancer relapse.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (T.S.); (S.D.)
| | - Sarah Bloukh
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Valerie J. Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Enas Alwohoush
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Jomana Bakeer
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Sarah Darwish
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (T.S.); (S.D.)
| | - Belal Azab
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Correspondence:
| |
Collapse
|
46
|
Studies of Non-Protective Autophagy Provide Evidence that Recovery from Therapy-Induced Senescence is Independent of Early Autophagy. Int J Mol Sci 2020; 21:ijms21041427. [PMID: 32093197 PMCID: PMC7073138 DOI: 10.3390/ijms21041427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 01/10/2023] Open
Abstract
Autophagy and senescence, predominant responses that may dictate cell fate after chemotherapy or radiation, often occur in tandem. Cells in states of senescence and/or autophagy are frequently growth arrested. We have previously reported that tumor cells induced into senescence by therapy can re-emerge from the growth-arrested state, a phenomenon termed proliferative recovery. The current work shows that, while tumor cells collaterally induced into senescence and autophagy by etoposide, doxorubicin, or radiation undergo proliferative recovery, neither pharmacological nor genetic inhibition of early autophagy alter the extent of senescence or the ability of cells to recover from senescence. These findings confirm and extend our previous observations, essentially dissociating senescence from autophagy, and further indicate that re-emergence from senescence does not appear to be facilitated by or dependent on autophagy. Our results also provide additional evidence for the promotion of the non-protective form of autophagy by both chemotherapeutic drugs and radiation, which may complicate current efforts to inhibit autophagy for therapeutic benefit.
Collapse
|
47
|
Wang Z, Gao J, Ohno Y, Liu H, Xu C. Rosiglitazone ameliorates senescence and promotes apoptosis in ovarian cancer induced by olaparib. Cancer Chemother Pharmacol 2020; 85:273-284. [PMID: 31907647 DOI: 10.1007/s00280-019-04025-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Senescence mechanisms are vital to resistance to long-term olaparib maintenance treatment. Recently, peroxisome proliferator-activated receptor-γ agonists (e.g., rosiglitazone) have been reported to ameliorate the senescence-like phenotype by modulating inflammatory mediator production. This study examined synergistic effects on the anti-tumor activity of rosiglitazone combined with olaparib in ovarian cancer treatment. METHODS A2780 and SKOV3 mouse subcutaneous xenograft models were established for observing anti-tumor effects in living organisms and were randomly split into combination (both olaparib and rosiglitazone), rosiglitazone (10 mg/kg), olaparib (10 mg/kg), control (solvent) groups that received treatment once every 2 or 3 days (n = 6 per group). Cell counting kit-8 (CCK-8) assays were used to test the influences of rosiglitazone and olaparib on cell proliferation. PI and Annexin-V-FITC staining was used with flow cytometry to assess the cell cycle distribution and cell apoptosis. Senescence-associated β-galactosidase (SA-β-Gal) staining was used to observe cellular senescence. We performed quantitative real-time polymerase chain reaction assays to study the senescence-related secretory phenotype (SASP). RESULTS Olaparib and rosiglitazone were observed to synergistically retard subcutaneous ovarian cancer growth in vivo, and synergistically suppress ovarian cancer cell proliferation in vitro. Compared with olaparib alone, the percentage of positive cells expressed SA-β-gal and SASP were significantly decreased in the treatment of combination of olaparib and rosiglitazone. Furthermore, olaparib plus rosiglitazone increased the percentage of apoptosis in ovarian cancer cell compared with olaparib alone. In A2780 cells, it showed lower expression of P53, phospho-p53 (Ser15), P21, and P18 protein in combination treatment compared with olaparib alone. While, in SKOV3 cells, it showed lower expression of phosphor-retinoblastoma protein (Rb) (Ser807/811), and higher expression of cyclin D1, P21, and P16 protein in combination treatment compared with olaparib alone. CONCLUSIONS Rosiglitazone combined with olaparib can help manage ovarian cancer by ameliorating olaparib-induced senescence and improving anti-tumor effects.
Collapse
Affiliation(s)
- Zehua Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Jianwen Gao
- Department of Health Science, Graduate School of Medical, Osaka University, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Major of Biotechnological Pharmaceutics, Shanghai Pharmaceutical School, Shanghai, 200135, China
| | - Yuko Ohno
- Department of Health Science, Graduate School of Medical, Osaka University, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Haiou Liu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China.
| | - Congjian Xu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China. .,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
48
|
Alotaibi MR, As Sobeai HM, Alaqil FA, Almutairi M, Alhazzani K, Sulaiman AA, Isab AA, Hadal Alotaibi N. A newly synthesized platinum-based compound (PBC-II) increases chemosensitivity of HeLa ovarian cancer cells via inhibition of autophagy. Saudi Pharm J 2019; 27:1203-1209. [PMID: 31885480 PMCID: PMC6921179 DOI: 10.1016/j.jsps.2019.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/02/2019] [Indexed: 11/11/2022] Open
Abstract
There are many mechanisms of resistance, chemoresistance of HeLa cells to anti-cancer agents seems to be autophagy-mediated. While using very effective anti-cancers such as Doxorubicin and cisplatin, cells overcome the cytotoxicity of these drugs through promotion of what so-called cytoprotective autophagy. Here in this study, we sought to introduce a novel platinum-based compound PBC-II that possesses anti-cancer activity. Our data showed that PBC-II is able to induce apoptosis at relatively low concentrations, with no detectable reactive oxygen species (ROS). However, further experiments demonstrated that exposure of HeLa cells to PBC-II did not promote autophagy; rather, it resulted in accumulation of p62 and decrease in LC3-II levels. Autophagy was then promoted in HeLa cells pharmacologically by Doxorubicin and genetically by siRNA IL-10. In order to confirm promotion of autophagy in our model, we performed acridine orange staining to assess for autophagy under microscope as well as via flow cytometry. We then measured protein level of autophagy markers p62 and LC3 by western blot. Our data indicated that PBC-II interferes with therapy-induced autophagy. We also determined PI3K activity while co-incubation of PBC-II with autophagy inducers. It was clear that PI3K activation decreased when PBC-II was co-administered with autophagy inducers. Collectively, PBC-II exerts unique anti-proliferative effects associated with inhibition of autophagy, which indicates that PBC-II is potentially a promising agent to be used in resistant ovarian tumors.
Collapse
Affiliation(s)
- Moureq Rashed Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Homood Moqbel As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | | | - Mashal Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Adam A.A. Sulaiman
- Lab Technical Support Office (LTSO), King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Anvarhusein A. Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Nasser Hadal Alotaibi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakakah 72341, Saudi Arabia
| |
Collapse
|
49
|
Wéra AC, Lobbens A, Stoyanov M, Lucas S, Michiels C. Radiation-induced synthetic lethality: combination of poly(ADP-ribose) polymerase and RAD51 inhibitors to sensitize cells to proton irradiation. Cell Cycle 2019; 18:1770-1783. [PMID: 31238782 DOI: 10.1080/15384101.2019.1632640] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Although improvements in radiation therapy were made over the years, radioresistance is still a major challenge. Cancer cells are often deficient for DNA repair response, a feature that is currently exploited as a new anti-cancer strategy. In this context, combination of inhibitors targeting complementary pathways is of interest to sensitize cells to radiation. In this work, we used PARP (Olaparib) and RAD51 (B02) inhibitors to radiosensitize cancer cells to proton and X-ray radiation. More particularly, Olaparib and B02 were used at concentration leading to limited cytotoxic (alone or in combination) but increasing cell death when the cells were irradiated. We showed that, although at limited concentration, Olaparib and B02 were able to radiosensitize different cancer cell lines, i.e. lung and pancreatic cancer cells. Antagonistic, additive or synergistic effects were observed and correlated to cell proliferation rate. The inhibitors enhanced persistent DNA damage, delayed apoptosis, prolonged cell cycle arrest and senescence upon irradiation. These results demonstrated that radiation-induced synthetic lethality might widen the therapeutic window, hence extending the use of PARP inhibitors to patients without BRCAness.
Collapse
|
50
|
Smigiel JM, Taylor SE, Bryson BL, Tamagno I, Polak K, Jackson MW. Cellular plasticity and metastasis in breast cancer: a pre- and post-malignant problem. JOURNAL OF CANCER METASTASIS AND TREATMENT 2019; 5:47. [PMID: 32355893 PMCID: PMC7192216 DOI: 10.20517/2394-4722.2019.26] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
As a field we have made tremendous strides in treating breast cancer, with a decline in the past 30 years of overall breast cancer mortality. However, this progress is met with little affect once the disease spreads beyond the primary site. With a 5-year survival rate of 22%, 10-year of 13%, for those patients with metastatic breast cancer (mBC), our ability to effectively treat wide spread disease is minimal. A major contributing factor to this ineffectiveness is the complex make-up, or heterogeneity, of the primary site. Within a primary tumor, secreted factors, malignant and pre-malignant epithelial cells, immune cells, stromal fibroblasts and many others all reside alongside each other creating a dynamic environment contributing to metastasis. Furthermore, heterogeneity contributes to our lack of understanding regarding the cells' remarkable ability to undergo epithelial/non-cancer stem cell (CSC) to mesenchymal/CSC (E-M/CSC) plasticity. The enhanced invasion & motility, tumor-initiating potential, and acquired therapeutic resistance which accompanies E-M/CSC plasticity implicates a significant role in metastasis. While most work trying to understand E-M/CSC plasticity has been done on malignant cells, recent evidence is emerging concerning the ability for pre-malignant cells to undergo E-M/CSC plasticity and contribute to the metastatic process. Here we will discuss the importance of E-M/CSC plasticity within malignant and pre-malignant populations of the tumor. Moreover, we will discuss how one may potentially target these populations, ultimately disrupting the metastatic cascade and increasing patient survival for those with mBC.
Collapse
Affiliation(s)
- Jacob M. Smigiel
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Sarah E. Taylor
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benjamin L. Bryson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Kelsey Polak
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mark W. Jackson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|