1
|
Paiboonborirak C, Abu-Rustum NR, Wilailak S. Artificial intelligence in the diagnosis and management of gynecologic cancer. Int J Gynaecol Obstet 2025. [PMID: 40277295 DOI: 10.1002/ijgo.70094] [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: 12/08/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 04/26/2025]
Abstract
Gynecologic cancers affect over 1.2 million women globally each year. Early diagnosis and effective treatment are essential for improving patient outcomes, yet traditional diagnostic methods often encounter limitations, particularly in low-resource settings. Artificial intelligence (AI) has emerged as a transformative tool that enhances accuracy and efficiency across various aspects of gynecologic oncology, including screening, diagnosis, and treatment. This review examines the current applications of AI in gynecologic cancer care, focusing on areas such as early detection, imaging, personalized treatment planning, and patient monitoring. Based on an analysis of 75 peer-reviewed articles published between 2017 and 2024, we highlight AI's contributions to cervical, ovarian, and endometrial cancer management. AI has notably improved early detection, achieving up to 95% accuracy in cervical cancer screening through AI-enhanced Pap smear analysis and colposcopy. For ovarian and endometrial cancers, AI-driven imaging and biomarker detection have enabled more personalized treatment approaches. In addition, AI tools have enhanced precision in robotic-assisted surgery and radiotherapy, and AI-based histopathology has reduced diagnostic variability. Despite these advancements, challenges such as data privacy, bias, and the need for human oversight must be addressed. The successful integration of AI into clinical practice will require careful consideration of ethical issues and a balanced approach that incorporates human expertise. Overall, AI presents significant potential to improve outcomes in gynecologic oncology, particularly in bridging healthcare gaps in resource-limited settings.
Collapse
Affiliation(s)
- Chaiyawut Paiboonborirak
- Department of Obstetrics and Gynecology, Bangkok Metropolitan Administration General Hospital (Klang Hospital), Bangkok, Thailand
| | - Nadeem R Abu-Rustum
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of OB/GYN, Weill Cornell Medical College, New York, New York, USA
| | - Sarikapan Wilailak
- Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Nair M, Samidurai A, Das A, Kakar SS, Kukreja RC. Ovarian cancer and the heart: pathophysiology, chemotherapy-induced cardiotoxicity, and new therapeutic strategies. J Ovarian Res 2025; 18:72. [PMID: 40188339 PMCID: PMC11971845 DOI: 10.1186/s13048-025-01636-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/21/2025] [Indexed: 04/07/2025] Open
Abstract
Ovarian Cancer (OC) is recognized as the most lethal gynecologic malignancy, characterized by numerous genetic mutations that trigger uncontrolled cellular growth and replication. Emerging evidence suggests that non-coding RNAs including miRNAs and lncRNAs significantly influence OC through their multiple roles including tumor initiation, progression, metastasis, immune evasion, and chemoresistance, making them promising diagnostic markers and therapeutic targets. The primary approach to treating OC typically involves cytoreductive surgery followed by chemotherapy. However, the chemotherapeutic agents, particularly the anthracyclines such as doxorubicin (DOX), are known for their cardiotoxic effects, which can range from acute to chronic, potentially leading to heart failure and death. To enhance the overall treatment response and to minimize cardiotoxicity, alternative strategies have been explored. These include the use of liposomal doxorubicin (DOXIL) as a substitute for DOX, various radiotherapies, immunotherapies, and the co-administration of angiotensin-converting enzyme inhibitors and/or beta-blockers. Phosphodiesterase-5 inhibitors (PDE5i) have also demonstrated efficacy in reducing cardiotoxicity linked to cancer treatments and in promoting apoptosis in cancer cells across multiple cancer types. Although there is no current clinical trial directly examining the impact of PDE5i on reducing cardiotoxicity in OC, however emerging therapies such as Withaferin A, PARP inhibitors, and nanoparticle combination therapy show promise. Additional research is essential to develop treatments that are both effective against OC and less harmful to the heart.
Collapse
Affiliation(s)
- Megha Nair
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Arun Samidurai
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Anindita Das
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Sham S Kakar
- Department of Physiology, University of Louisville, Louisville, KY, USA
| | - Rakesh C Kukreja
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
| |
Collapse
|
3
|
Petty M, Jhaveri V, Taylor N, Foster B. Oligometastatic granulosa cell tumor treated with stereotactic body radiation therapy with a radiographic and tumor marker response. Gynecol Oncol Rep 2025; 58:101708. [PMID: 40103618 PMCID: PMC11914764 DOI: 10.1016/j.gore.2025.101708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Stereotactic body radiation therapy (SBRT) is a safe and effective treatment for oligometastatic gynecologic cancers. This report details the case of a 75-year-old woman with recurrent granulosa cell tumor of the left ovary. The patient was diagnosed with a peritoneal recurrence after her inhibin-B level increased to 39.7 pg/mL, and a subsequent computed tomography (CT) scan of the abdomen and pelvis revealed an enlarging left-sided soft tissue mass in the sigmoid mesentery measuring 1.2 × 1.2 cm. She underwent SBRT, delivering 37.5 Gy in 5 fractions to the soft tissue mass. A CT scan post-treatment indicated the mass had decreased to 0.8 cm and that her inhibin B level decreased to < 7 pg/mL. This case highlights the effectiveness of SBRT in the management of oligometastatic ovarian cancer, achieving a marked radiographic and tumor marker response in patients with disease not amenable to surgical resection.
Collapse
Affiliation(s)
- Madeline Petty
- Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA 19140, United States
| | - Vasanti Jhaveri
- Department of Gynecology Oncology, St. Luke's University Health Network, 701 Ostrum St., 5th Floor, Bethlehem, PA 18015, United States
| | - Nicholas Taylor
- Department of Gynecology Oncology, St. Luke's University Health Network, 701 Ostrum St., 5th Floor, Bethlehem, PA 18015, United States
| | - Benjamin Foster
- Department of Radiation Oncology, St. Luke's University Health Network, 801 Ostrum St., Bethlehem, PA 18015, United States
| |
Collapse
|
4
|
Durante S, Cuccia F, Rigo M, Caminiti G, Mastroleo F, Lazzari R, Corrao G, Caruso G, Vigorito S, Cattani F, Ferrera G, Chiantera V, Alongi F, Colombo N, Jereczek-Fossa BA. Stereotactic radiotherapy for managing ovarian cancer oligoprogression under poly (ADP-ribose) polymerase inhibitors (PARPi). Int J Gynecol Cancer 2024; 34:1232-1239. [PMID: 38821546 DOI: 10.1136/ijgc-2024-005361] [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: 02/06/2024] [Accepted: 05/20/2024] [Indexed: 06/02/2024] Open
Abstract
OBJECTIVE Poly (ADP-ribose) polymerase inhibitors (PARPi) have become a new standard of care for the maintenance treatment of advanced epithelial ovarian cancer. This study aims to evaluate the efficacy and safety of combining stereotactic body radiotherapy with PARPi continuation as a strategy to treat ovarian cancer oligoprogression on PARPi. METHODS This is a multicenter retrospective study including ovarian cancer patients treated with stereotactic body radiotherapy and PARPi continuation for oligoprogression under PARPi maintenance therapy between June 2012 and May 2023 in three Italian centers. PARPi treatment was continued until further disease progression or unacceptable toxicity. The primary endpoint was the next-line systemic therapy-free interval. The Kaplan-Meier method was used to assess local control, progression-free survival, and overall survival. Univariate and multivariate Cox regression analyses were performed to evaluate potential clinical outcomes predictors. RESULTS 46 patients were included, with a total of 89 lesions treated over 63 radiotherapy treatments. Lymph nodes were the most frequently treated lesions (80, 89.9%), followed by visceral lesions (8, 9%) and one case with a bone lesion (1.1%). Median follow-up was 25.9 months (range 2.8-122). The median next-line systemic therapy-free interval was 12.4 months (95% CI 8.3 to 19.5). A number of prior chemotherapy lines greater than five was significantly associated with a reduced next-line systemic therapy-free interval (HR 3.21, 95% CI 1.11 to 9.32, p=0.032). At the time of analysis, 32 (69.6%) patients started a new systemic therapy regimen, while 14 (30.4%) remained on the PARPi regimen. The 2-year progression-free survival, local failure-free survival, and overall survival rates were 10.7%, 78.1%, and 76.5%, respectively. Four patients (8.7%) experienced acute toxicity with G1 gastrointestinal events. CONCLUSION Stereotactic body radiotherapy combined with PARPi continuation may be an effective and safe strategy for managing ovarian cancer patients with oligoprogression on PARPi maintenance therapy. Prospective research is warranted to shed more light on this approach.
Collapse
Affiliation(s)
- Stefano Durante
- Division of Radiation Oncology, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | | | - Michele Rigo
- Advanced Radiation Oncology Department, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar, Veneto, Italy
| | | | - Federico Mastroleo
- Division of Radiation Oncology, European Institute of Oncology, IEO, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Roberta Lazzari
- Division of Radiation Oncology, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Giulia Corrao
- Division of Radiation Oncology, European Institute of Oncology, IEO, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Caruso
- Department of Experimental Medicine, University of Rome La Sapienza, Rome, Lazio, Italy
- Divison of Gynecologic Oncology, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Sabrina Vigorito
- Unit of Medical Physics, European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Cattani
- Unit of Medical Physics, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Vito Chiantera
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Sicilia, Italy
- Department of Gynecologic Oncology, Istituto Nazionale Tumori - IRCCS Fondazione G. Pascale, Naples, Italy
| | - Filippo Alongi
- Advanced Radiation Oncology Department, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar, Veneto, Italy
- University of Brescia, Brescia, Italy
| | - Nicoletta Colombo
- Gynecology Program, European Institute of Oncology, IEO, IRCCS, Milan, Italy
- Department of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, European Institute of Oncology, IEO, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
5
|
Kou X, Yang X, Zhao Z, Li L. HSPA8-mediated stability of the CLPP protein regulates mitochondrial autophagy in cisplatin-resistant ovarian cancer cells. Acta Biochim Biophys Sin (Shanghai) 2024; 56:356-365. [PMID: 38419499 PMCID: PMC10984867 DOI: 10.3724/abbs.2023246] [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: 03/17/2023] [Accepted: 08/07/2023] [Indexed: 03/02/2024] Open
Abstract
Currently, platinum agents remain the mainstay of chemotherapy for ovarian cancer (OC). However, cisplatin (DDP) resistance is a major reason for chemotherapy failure. Thus, it is extremely important to elucidate the mechanism of resistance to DDP. Here, we establish two DDP-resistant ovarian cancer cell lines and find that caseinolytic protease P (CLPP) level is significantly downregulated in DDP-resistant cell lines compared to wild-type ovarian cancer cell lines (SK-OV-3 and OVcar3). Next, we investigate the functions of CLPP in DDP-resistant and wild-type ovarian cancer cells using various assays, including cell counting kit-8 assay, western blot analysis, immunofluorescence staining, and detection of reactive oxygen species (ROS) and apoptosis. Our results show that CLPP knockdown significantly increases the half maximal inhibitory concentration (IC 50) and mitophagy of wild-type SK-OV-3 and OVcar3 cells, while CLPP overexpression reduces the IC 50 values and mitophagy of DDP-resistant SK-OV-3 and OVcar3 cells. Next, we perform database predictions and confirmation experiments, which show that heat shock protein family A member 8 (HSPA8) regulates CLPP protein stability. The dynamic effects of the HSPA8/CLPP axis in ovarian cancer cells are also examined. HSPA8 increases mitophagy and the IC 50 values of SK-OV-3 and OVcar3 cells but inhibits their ROS production and apoptosis. In addition, CLPP partly reverses the effects induced by HSPA8 in SK-OV-3 and OVcar3 cells. In conclusion, CLPP increases DDP resistance in ovarian cancer by inhibiting mitophagy and promoting cellular stress. Meanwhile, HSPA8 promotes the degradation of CLPP protein by regulating its stability.
Collapse
Affiliation(s)
- Xinxin Kou
- />Department of GynecologyCancer Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| | - Xiaoxia Yang
- />Department of GynecologyCancer Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| | - Zheng Zhao
- />Department of GynecologyCancer Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| | - Lei Li
- />Department of GynecologyCancer Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| |
Collapse
|
6
|
Hamade DF, Epperly MW, Fisher R, Hou W, Shields D, van Pijkeren JP, Leibowitz BJ, Coffman LG, Wang H, Huq MS, Huang Z, Rogers CJ, Vlad AM, Greenberger JS, Mukherjee A. Genetically Engineered Probiotic Limosilactobacillus reuteri Releasing IL-22 (LR-IL-22) Modifies the Tumor Microenvironment, Enabling Irradiation in Ovarian Cancer. Cancers (Basel) 2024; 16:474. [PMID: 38339228 PMCID: PMC10854600 DOI: 10.3390/cancers16030474] [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: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Despite recent advances in cancer therapy, ovarian cancer remains the most lethal gynecological cancer worldwide, making it crucial and of the utmost importance to establish novel therapeutic strategies. Adjuvant radiotherapy has been assessed historically, but its use was limited by intestinal toxicity. We recently established the role of Limosilactobacillus reuteri in releasing IL-22 (LR-IL-22) as an effective radiation mitigator, and we have now assessed its effect in an ovarian cancer mouse model. We hypothesized that an LR-IL-22 gavage would enable intestinal radioprotection by modifying the tumor microenvironment and, subsequently, improving overall survival in female C57BL/6MUC-1 mice with widespread abdominal syngeneic 2F8cis ovarian cancer. Herein, we report that the LR-IL-22 gavage not only improved overall survival in mice when combined with a PD-L1 inhibitor by inducing differential gene expression in irradiated stem cells but also induced PD-L1 protein expression in ovarian cancer cells and mobilized CD8+ T cells in whole abdomen irradiated mice. The addition of LR-IL-22 to a combined treatment modality with fractionated whole abdomen radiation (WAI) and systemic chemotherapy and immunotherapy regimens can facilitate a safe and effective protocol to reduce tumor burden, increase survival, and improve the quality of life of a locally advanced ovarian cancer patient.
Collapse
Affiliation(s)
- Diala F. Hamade
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | | | - Brian J. Leibowitz
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Lan G. Coffman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.W.); (Z.H.)
| | - M. Saiful Huq
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Ziyu Huang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.W.); (Z.H.)
| | | | - Anda M. Vlad
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20850, USA;
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| |
Collapse
|
7
|
Shen J, Tao Y, Zhou J, Guan H, Zhen H, Yan J, Hou X, Liu Z, Hu K, Zhang F. Is Prophylactic Radiotherapy to the Lymphatic Drainage Area Necessary for Patients With Stage III Ovarian Cancer After Chemotherapy Following Surgery? Cancer Control 2024; 31:10732748241263703. [PMID: 38907367 PMCID: PMC11193923 DOI: 10.1177/10732748241263703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 05/15/2024] [Accepted: 05/31/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND For patients with stage III epithelial ovarian cancer, there are limited studies on the effects of postoperative adjuvant radiotherapy (RT). Here we assessed the therapeutic efficacy and toxicity of postoperative radiotherapy to the abdominal and pelvic lymphatic drainage area for stage III epithelial ovarian cancer patients, who had all received surgery and chemotherapy (CT). METHODS We retrospectively collected patients with stage III epithelial ovarian cancer after cytoreductive surgery (CRS) and full-course adjuvant CT. The chemoradiotherapy (CRT) group patients were treated with intensity modulated radiotherapy (IMRT) to the abdominal and pelvic lymphatic drainage area in our hospital between 2010 and 2020. A propensity score matching analysis was conducted to compare the results between the CRT and CT groups. Kaplan-Meier analysis estimated overall survival (OS), disease-free survival (DFS), and local control (LC) rates. The log-rank test determined the significance of prognostic factors. RESULTS A total of 132 patients with median follow-up of 73.9 months (9.1-137.7 months) were included (44 and 88 for the CRT and RT groups, retrospectively). The baseline characteristics of age, histology, level of CA12-5, surgical staging, residual tumour, courses of adjuvant CT, and courses to reduce CA12-5 to normal were all balanced. The median DFS time, 5-year OS, and local recurrence free survival (LRFS) were 100.0 months vs 25.9 months (P = .020), 69.2% vs 49.9% (P = .002), and 85.9% vs 50.5% (P = .020), respectively. The CRT group mainly presented with acute haematological toxicities, with no statistically significant difference compared with grade III intestinal adverse effects (3/44 vs 6/88, P = .480). CONCLUSION This report demonstrates that long-term DFS could be achieved in stage III epithelial ovarian cancer patients treated with IMRT preventive radiation to the abdominal and pelvic lymphatic area. Compared with the CT group, DFS and OS were significantly prolonged and adverse effects were acceptable.
Collapse
Affiliation(s)
- Jing Shen
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Yinjie Tao
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Jingya Zhou
- Department of medical record, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Hui Guan
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Hongnan Zhen
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Junfang Yan
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Xiaorong Hou
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Zhikai Liu
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Ke Hu
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| | - Fuquan Zhang
- Department of radiation oncology, Peking Union Medical College Hospital, Dongcheng District, Beijing, People’s Republic of China
| |
Collapse
|
8
|
Macchia G, Titone F, Restaino S, Arcieri M, Pellecchia G, Andreetta C, Driul L, Vizzielli G, Pezzulla D. Is It Time to Reassess the Role of Radiotherapy Treatment in Ovarian Cancer? Healthcare (Basel) 2023; 11:2413. [PMID: 37685447 PMCID: PMC10486999 DOI: 10.3390/healthcare11172413] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
With a 5-year survival rate of fewer than 50%, epithelial ovarian carcinoma is the most fatal of the gynecologic cancers. Each year, an estimated 22,000 women are diagnosed with the condition, with 14,000 dying as a result, in the United States. Over the last decade, the advent of molecular and genetic data has enhanced our understanding of the heterogeneity of ovarian cancer. More than 80% of women diagnosed with advanced illness have an initial full response to rigorous therapy at diagnosis, including surgery and platinum-based chemotherapy. Unfortunately, these responses are infrequently lasting, and the majority of women with ovarian cancer suffer recurrent disease, which is often incurable, despite the possibility of future response and months of survival. And what therapeutic weapons do we have to counter it? For many years, radiation therapy for ovarian tumors was disregarded as an effective treatment option due to its toxicity and lack of survival benefits. Chemotherapy is widely used following surgery, and it has nearly completely supplanted radiation therapy. Even with the use of more modern and efficient chemotherapy regimens, ovarian cancer failures still happen. After receiving first-line ovarian cancer chemotherapy, over 70% of patients show evidence of recurrence in the abdomen or pelvis. It is necessary to reinterpret the function of radiation therapy in light of recent technological developments, the sophistication of radiation procedures, and the molecular and biological understanding of various histological subtypes. This review article focuses on the literature on the use of radiation in ovarian tumors as well as its rationale and current indications.
Collapse
Affiliation(s)
- Gabriella Macchia
- Radiation Oncology Unit, Responsible Research Hospital, 86100 Campobasso, Italy; (G.M.); (D.P.)
| | - Francesca Titone
- Radiation Oncology Unit, Department of Oncology, “Santa Maria della Misericordia” University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
| | - Stefano Restaino
- Department of Maternal and Child Health, “Santa Maria della Misericordia” University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (S.R.); (M.A.); (G.P.); (L.D.); (G.V.)
| | - Martina Arcieri
- Department of Maternal and Child Health, “Santa Maria della Misericordia” University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (S.R.); (M.A.); (G.P.); (L.D.); (G.V.)
- Department of Biomedical, Dental, Morphological and Functional Imaging Science, University of Messina, 98122 Messina, Italy
| | - Giulia Pellecchia
- Department of Maternal and Child Health, “Santa Maria della Misericordia” University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (S.R.); (M.A.); (G.P.); (L.D.); (G.V.)
- Medical Area Department (DAME), University of Udine, 33100 Udine, Italy
| | - Claudia Andreetta
- Medical Oncology Unit, Department of Oncology, “Santa Maria della Misericordia” University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy;
| | - Lorenza Driul
- Department of Maternal and Child Health, “Santa Maria della Misericordia” University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (S.R.); (M.A.); (G.P.); (L.D.); (G.V.)
- Medical Area Department (DAME), University of Udine, 33100 Udine, Italy
| | - Giuseppe Vizzielli
- Department of Maternal and Child Health, “Santa Maria della Misericordia” University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy; (S.R.); (M.A.); (G.P.); (L.D.); (G.V.)
- Medical Area Department (DAME), University of Udine, 33100 Udine, Italy
| | - Donato Pezzulla
- Radiation Oncology Unit, Responsible Research Hospital, 86100 Campobasso, Italy; (G.M.); (D.P.)
| |
Collapse
|
9
|
Sproull M, Wilson E, Miller R, Camphausen K. The Future of Radioactive Medicine. Radiat Res 2023; 200:80-91. [PMID: 37141143 PMCID: PMC10466314 DOI: 10.1667/rade-23-00031.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/07/2023] [Indexed: 05/05/2023]
Abstract
The discovery of X rays in the late 19th century heralded the beginning of a new age in medicine, and the advent of channeling the power of radiation to diagnose and treat human disease. Radiation has been leveraged in medicine in a multitude of ways and is a critical element of cancer care including screening, diagnosis, surveillance, and interventional treatments. Modern radiotherapy techniques include a multitude of methodologies utilizing both externally and internally delivered radiation from a variety of approaches. This review provides a comprehensive overview of contemporary radiotherapy methodologies, the field of radiopharmaceuticals and theranostics, effects of low dose radiation and highlights the phenomena of fear of exposure to radiation and its impact in modern medicine.
Collapse
Affiliation(s)
- M. Sproull
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - E. Wilson
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - R.W. Miller
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - K. Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
10
|
Zhao X, Liu Z, Chen T. Potential Role of Vaginal Microbiota in Ovarian Cancer Carcinogenesis, Progression and Treatment. Pharmaceutics 2023; 15:pharmaceutics15030948. [PMID: 36986809 PMCID: PMC10056320 DOI: 10.3390/pharmaceutics15030948] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Ovarian cancer represents one of the most challenging gynecologic cancers which still has numerous unknowns on the underlying pathogenesis. In addition to the verified contributors such as genomic predisposition and medical history in the carcinogenesis, emerging evidence points out the potential role of vaginal microbiota in ovarian cancer. Recent studies have underlined the presence of vaginal microbial dysbiosis in cancer cases. Increasing research also indicates the potential correlations between vaginal microbes and cancer carcinogenesis, progression and treatment. Currently, compared with other gynecologic cancers, reports on the roles of vaginal microbiota in ovarian cancer remain scarce and fragmentary. Therefore, in this review, we summarize the roles of vaginal microbiota in various gynecologic diseases, particularly focusing on the potential mechanisms and possible applications of vaginal microbiota in ovarian cancer, giving insight into the involvement of vaginal microbiota in gynecologic cancer treatment.
Collapse
Affiliation(s)
- Xiumiao Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Correspondence: (Z.L.); (T.C.)
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- National Engineering Research Center for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
- Correspondence: (Z.L.); (T.C.)
| |
Collapse
|
11
|
Varier L, Sundaram SM, Gamit N, Warrier S. An Overview of Ovarian Cancer: The Role of Cancer Stem Cells in Chemoresistance and a Precision Medicine Approach Targeting the Wnt Pathway with the Antagonist sFRP4. Cancers (Basel) 2023; 15:cancers15041275. [PMID: 36831617 PMCID: PMC9954718 DOI: 10.3390/cancers15041275] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Ovarian cancer is one of the most prevalent gynecological cancers, having a relatively high fatality rate with a low five-year chance of survival when detected in late stages. The early detection, treatment and prevention of metastasis is pertinent and a pressing research priority as many patients are diagnosed only in stage three of ovarian cancer. Despite surgical interventions, targeted immunotherapy and adjuvant chemotherapy, relapses are significantly higher than other cancers, suggesting the dire need to identify the root cause of metastasis and relapse and present more precise therapeutic options. In this review, we first describe types of ovarian cancers, the existing markers and treatment modalities. As ovarian cancer is driven and sustained by an elusive and highly chemoresistant population of cancer stem cells (CSCs), their role and the associated signature markers are exhaustively discussed. Non-invasive diagnostic markers, which can be identified early in the disease using circulating tumor cells (CTCs), are also described. The mechanism of the self-renewal, chemoresistance and metastasis of ovarian CSCs is regulated by the Wnt signaling pathway. Thus, its role in ovarian cancer in promoting stemness and metastasis is delineated. Based on our findings, we propose a novel strategy of Wnt inhibition using a well-known Wnt antagonist, secreted frizzled related protein 4 (sFRP4), wherein short micropeptides derived from the whole protein can be used as powerful inhibitors. The latest approaches to early diagnosis and novel treatment strategies emphasized in this review will help design precision medicine approaches for an effective capture and destruction of highly aggressive ovarian cancer.
Collapse
Affiliation(s)
- Lavanya Varier
- Cuor Stem Cellutions Pvt Ltd., Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | - S. Mohana Sundaram
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | - Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | - Sudha Warrier
- Cuor Stem Cellutions Pvt Ltd., Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
- Correspondence:
| |
Collapse
|
12
|
Kim TW, Lee HG. 6-Shogaol Overcomes Gefitinib Resistance via ER Stress in Ovarian Cancer Cells. Int J Mol Sci 2023; 24:ijms24032639. [PMID: 36768961 PMCID: PMC9916959 DOI: 10.3390/ijms24032639] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
In women, ovary cancer is already the fifth leading cause of mortality worldwide. The use of cancer therapies, such as surgery, radiotherapy, and chemotherapy, may be a powerful anti-cancer therapeutic strategy; however, these therapies still have many problems, including resistance, toxicity, and side effects. Therefore, natural herbal medicine has the potential to be used for cancer therapy because of its low toxicity, fewer side effects, and high success. This study aimed to investigate the anti-cancer effect of 6-shogaol in ovarian cancer cells. 6-shogaol induces ER stress and cell death via the reduction in cell viability, the increase in LDH cytotoxicity, caspase-3 activity, and Ca2+ release, and the upregulation of GRP78, p-PERK, p-eIF2α, ATF-4, CHOP, and DR5. Moreover, 6-shogaol treatment medicates endoplasmic reticulum (ER) stress and cell death by upregulating Nox4 and releasing ROS. The knockdown of Nox4 in ovarian cancer cells inhibits ER stress and cell death by blocking the reduction in cell viability and the enhancement of LDH cytotoxicity, caspase-3 activity, Ca2+, and ROS release. In gefitinib-resistant ovarian cancer cells, A2780R and OVCAR-3R, 6-shogaol/gefitinib overcomes gefitinib resistance by inhibiting EMT phenomena such as the reduction in E-cadherin, and the increase in N-cadherin, vimentin, Slug, and Snail. Therefore, our results suggest that 6-shogaol exerts a potential anti-cancer effect in ovarian cancer and combination treatment with 6-shogaol and gefitinib may provide a novel anti-tumor therapeutic strategy in gefitinib-resistant ovarian cancer.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, Gyeongju 38066, Republic of Korea
- Correspondence:
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
13
|
Bae BK, Cho WK, Lee JW, Kim TJ, Choi CH, Lee YY, Park W. Role of salvage radiotherapy for recurrent ovarian cancer. Int J Gynecol Cancer 2023; 33:66-73. [PMID: 36137577 DOI: 10.1136/ijgc-2022-003834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE This study aimed to report clinical outcomes of salvage radiotherapy for recurrent ovarian cancer and identify predictors of clinical outcomes. METHODS We retrospectively reviewed data of patients who received salvage radiotherapy for recurrent ovarian cancer between January 2011 and June 2021. Stereotactic body radiotherapy, involved-field radiotherapy with conventional fractionation, and non-involved-field radiotherapy with conventional fractionation were included in this study. Local failure-free survival, progression-free survival, chemotherapy-free survival, and overall survival were assessed. Additionally, potential prognostic factors for survival were analyzed. RESULTS A total of 79 patients were included with 114 recurrent lesions. The median follow-up was 18.3 months (range 1.7-83). The 2-year local failure-free survival, progression-free survival, chemotherapy-free survival, and overall survival rates were 80.7%, 10.6%, 21.2%, and 74.7%, respectively. Pre-radiotherapy platinum resistance (hazard ratio (HR) 3.326, p<0.001) and short pre-radiotherapy CA-125 doubling time (HR 3.664, p<0.001) were associated with poor chemotherapy-free survival. The 1-year chemotherapy-free survival rates of patients with both risk factors, a single risk factor, and no risk factor were 0%, 20.4%, and 53.5%, respectively. The difference between risk groups was statistically significant: low risk versus intermediate risk (p<0.001) and intermediate risk versus high risk (p<0.001). CONCLUSIONS Salvage radiotherapy for recurrent ovarian cancer resulted in local control with improved chemotherapy-free survival in carefully selected patients. Our results suggest that the consideration of pre-radiotherapy platinum resistance and pre-radiotherapy CA-125 doubling time could help with patient selection.
Collapse
Affiliation(s)
- Bong Kyung Bae
- Department of Radiation Oncology, Samsung Medical Center, Gangnam-gu, Seoul, Korea (Republic of)
| | - Won Kyung Cho
- Department of Radiation Oncology, Samsung Medical Center, Gangnam-gu, Seoul, Korea (Republic of)
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Gangnam-gu, Seoul, Korea (Republic of)
| | - Tae-Joong Kim
- Department of Obstetrics and Gynecology, Samsung Medical Center, Gangnam-gu, Seoul, Korea (Republic of)
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Gangnam-gu, Seoul, Korea (Republic of)
| | - Yoo-Young Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Gangnam-gu, Seoul, Korea (Republic of)
| | - Won Park
- Department of Radiation Oncology, Samsung Medical Center, Gangnam-gu, Seoul, Korea (Republic of)
| |
Collapse
|
14
|
Pangath M, Unnikrishnan L, Throwba PH, Vasudevan K, Jayaraman S, Li M, Iyaswamy A, Palaniyandi K, Gnanasampanthapandian D. The Epigenetic Correlation among Ovarian Cancer, Endometriosis and PCOS: A Review. Crit Rev Oncol Hematol 2022; 180:103852. [DOI: 10.1016/j.critrevonc.2022.103852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/08/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
|
15
|
Wang YC, Tian JY, Han YY, Liu YF, Chen SY, Guo FJ. Evaluation of the potential of ultrasound-mediated drug delivery for the treatment of ovarian cancer through preclinical studies. Front Oncol 2022; 12:978603. [PMID: 36132133 PMCID: PMC9483181 DOI: 10.3389/fonc.2022.978603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian cancer (OC) has the greatest mortality rate among gynecological cancers, with a five-year survival rate of <50%. Contemporary adjuvant chemotherapy mostly fails in the case of OCs that are refractory, metastatic, recurrent, and drug-resistant. Emerging ultrasound (US)-mediated technologies show remarkable promise in overcoming these challenges. Absorption of US waves by the tissue results in the generation of heat due to its thermal effect causing increased diffusion of drugs from the carriers and triggering sonoporation by increasing the permeability of the cancer cells. Certain frequencies of US waves could also produce a cavitation effect on drug-filled microbubbles (MBs, phospholipid bilayers) thereby generating shear force and acoustic streaming that could assist drug release from the MBs, and promote the permeability of the cell membrane. A new class of nanoparticles that carry therapeutic agents and are guided by US contrast agents for precision delivery to the site of the ovarian tumor has been developed. Phase-shifting of nanoparticles by US sonication has also been engineered to enhance the drug delivery to the ovarian tumor site. These technologies have been used for targeting the ovarian cancer stem cells and protein moieties that are particularly elevated in OCs including luteinizing hormone-releasing hormone, folic acid receptor, and vascular endothelial growth factor. When compared to healthy ovarian tissue, the homeostatic parameters at the tissue microenvironment including pH, oxygen levels, and glucose metabolism differ significantly in ovarian tumors. US-based technologies have been developed to take advantage of these tumor-specific alterations for precision drug delivery. Preclinical efficacy of US-based targeting of currently used clinical chemotherapies presented in this review has the potential for rapid human translation, especially for formulations that use all substances that are deemed to be generally safe by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Yi-Chao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Jing-Yan Tian
- Department of Urology, The Second Division of the First Hospital of Jilin University, Changchun, China
| | - Ying-Ying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yun-Fei Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Si-Yao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Feng-Jun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Feng-Jun Guo,
| |
Collapse
|
16
|
Rehman U, Parveen N, Sheikh A, Abourehab MAS, Sahebkar A, Kesharwani P. Polymeric nanoparticles-siRNA as an emerging nano-polyplexes against ovarian cancer. Colloids Surf B Biointerfaces 2022; 218:112766. [PMID: 35994990 DOI: 10.1016/j.colsurfb.2022.112766] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Ovarian cancer (OC) is considered fifth-deadliest cancer globally responsible for high mortality in women. As the conventional therapeutic and diagnostic approaches are ineffective in increasing the survival rates of advanced staged patients by more than 5 years, OC has resulted in high morbidity and mortality rates over the last two decades. As a result, there is a dire need for innovative treatment approaches to address the issues. RNAi and nanotechnology can be considered the most appropriate strategies that can be used to improve OC therapy and help circumvent the chemo-resistance. siRNA is considered highly successful in facilitating the knockdown of specific genes on entering the cytosol when administered in-vivo via inhibiting the mRNA expression responsible for translation of those specific genes through the mechanism called RNA interference (RNAi). However, the primary barrier of utmost importance in the clinical efficacy of employed siRNA for the treatment of OC is the systemic distribution to the targeted site from the administration site. As a result, nanoparticles are constructed to carry the siRNA molecules inside them to the targeted site by preventing serum degradation and enhancing the serum stability of administered siRNA. The present review assesses the developments made in the polymeric-based nanoparticle siRNA delivery for targeting particular genes involved in the prognosis of ovarian cancers and surpassing the chemo-resistance and thus improving the therapeutic potentials of administered agents.
Collapse
Affiliation(s)
- Urushi Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Neha Parveen
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia 61519, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
17
|
Effects of Laparoscopic Hyperthermic Perfusion Therapy Combined with Adjuvant Treatment of Compound Yew Capsule on Ovarian Blood Flow Parameters and Immune Function in Patients with Ovarian Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9603492. [PMID: 35873625 PMCID: PMC9300267 DOI: 10.1155/2022/9603492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 11/28/2022]
Abstract
Objective To determine the effects of laparoscopic hyperthermic perfusion therapy combined with adjuvant compound yew capsules on ovarian blood flow parameters and immune function in patients with ovarian cancer (OC). Methods A total of 90 OC patients enrolled in our hospital between January 2019 and January 2020 were randomly distributed into the control (Con group) and experimental group (Exp group) based on the sealed envelope method. The Con group was administered laparoscopic hyperthermic perfusion therapy. On this basis, the Exp group was subjected to compound yew capsules; the ovarian blood flow parameters and immune function indexes were compared between the two groups. Results The Exp group was reported to perform better than the Con group regarding ovarian blood flow parameters and immune indexes after treatment (p < 0.001). Conclusion Laparoscopic hyperthermic perfusion therapy combined with adjuvant compound yew capsules for patients with OC can substantially improve the clinical indexes and immune function. Furthermore, research and adequate promotion are needed to elicit the evidence beyond preclinical studies to understand the intricacies of its implementation.
Collapse
|
18
|
Patra A, Nag A, Chakraborty A, Bhattacharyya N. PA1 cells containing a truncated DNA polymerase β protein are more sensitive to gamma radiation. Radiat Oncol J 2022; 40:66-78. [PMID: 35368202 PMCID: PMC8984132 DOI: 10.3857/roj.2021.00689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/20/2021] [Accepted: 01/05/2022] [Indexed: 11/03/2022] Open
Abstract
PURPOSE DNA polymerase β (Polβ) acts in the base excision repair (BER) pathway. Mutations in DNA polymerase β (Polβ) are associated with different cancers. A variant of Polβ with a 97 amino acid deletion (PolβΔ), in heterozygous conditions with wild-type Polβ, was identified in sporadic ovarian tumor samples. This study aims to evaluate the gamma radiation sensitivity of PolβΔ for possible target therapy in ovarian cancer treatment. MATERIALS AND METHODS PolβΔ cDNA was cloned in a GFP vector and transfected in PA1 cells. Stable cells (PA1PolβΔ) were treated with 60Co sourced gamma-ray (0-15 Gy) to investigate their radiation sensitivity. The affinity of PolβΔ with DNA evaluated by DNA protein in silico docking experiments. RESULTS The result showed a statistically significant (p < 0.05) higher sensitivity towards radiation at different doses (0-15 Gy) and time-point (48-72 hours) for PA1PolβΔ cells in comparison with normal PA1 cells. Ten Gy of gamma radiation was found to be the optimal dose. Significantly more PA1PolβΔ cells were killed at this dose than PA1 cells after 48 hours of treatment via an apoptotic pathway. The in silico docking experiments revealed that PolβΔ has more substantial binding potential towards the dsDNA than wild-type Polβ, suggesting a possible failure of BER pathway that results in cell death. CONCLUSION Our study showed that the PA1PolβΔ cells were more susceptible than PA1 cells to gamma radiation. In the future, the potentiality of ionizing radiation to treat this type of cancer will be checked in animal models.
Collapse
Affiliation(s)
- Anutosh Patra
- Department of Biotechnology, Panskura Banamali College, West Bengal, India
| | - Anish Nag
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, India
| | | | | |
Collapse
|
19
|
Chen JP, Shen CW, Hsueh FJ, Leung HC. Skin metastasis of a p16-positive squamous cell carcinoma mimicking radiation recall dermatitis. JOURNAL OF CANCER RESEARCH AND PRACTICE 2022. [DOI: 10.4103/jcrp.jcrp_28_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
20
|
Gan J, Herzog J, Smith DA, Vos D, Kikano E, Tirumani SH, Ramaiya NH. Primary peritoneal serous carcinoma: a primer for radiologists. Clin Imaging 2021; 83:56-64. [PMID: 34974267 DOI: 10.1016/j.clinimag.2021.12.007] [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: 08/07/2021] [Revised: 11/14/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022]
Abstract
Primary peritoneal serous carcinoma (PPSC) is a rare primary peritoneal tumor characterized by a unique range of clinical features and imaging findings. Though it shares many clinical, histologic, and imaging features with serous ovarian carcinoma, it remains a distinct clinical entity. Although less common than its primary ovarian counterpart, PPSC is characterized by a prognosis that is often equally poor with presentations common in late stages of disease. Key imaging modalities used in the evaluation of PPSC include ultrasound, CT, MRI, and PET/CT. For radiologists, an understanding of the pertinent imaging findings, pathologic correlations, and clinical features of PPSC is essential for arriving at the correct diagnosis and guiding the subsequent appropriate management of this complex malignancy.
Collapse
Affiliation(s)
- Jonathan Gan
- Case Western Reserve University, School of Medicine, 9501 Euclid Ave, Cleveland, OH 44106, United States of America
| | - Jackson Herzog
- Department of Radiology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, United States of America
| | - Daniel A Smith
- Department of Radiology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, United States of America.
| | - Derek Vos
- Case Western Reserve University, School of Medicine, 9501 Euclid Ave, Cleveland, OH 44106, United States of America
| | - Elias Kikano
- Department of Radiology, Brigham & Women's Hospital, 75 Francis St, Boston, MA 02115, United States of America
| | - Sree H Tirumani
- Department of Radiology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, United States of America
| | - Nikhil H Ramaiya
- Department of Radiology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, United States of America
| |
Collapse
|
21
|
Li D, Zhang M, Chao H. Significance of glutathione peroxidase 4 and intracellular iron level in ovarian cancer cells-"utilization" of ferroptosis mechanism. Inflamm Res 2021; 70:1177-1189. [PMID: 34537856 DOI: 10.1007/s00011-021-01495-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE AND DESIGN Ovarian cancer is the major cause of death in gynecologic diseases worldwide. Ferroptosis, a nonapoptotic form of cell death, is featured by accumulation of iron-based lipid peroxidation. The elevated iron level and malondialdehyde (MDA) in ovarian cancer cells suggest more vulnerable to ferroptosis, nevertheless, ferroptosis is not observed in ovarian cancer cells. Glutathione peroxidase 4 (GPX4) is a critical regulator of ferroptosis. METHODS We determined whether GPX4 knockdown could induce ferroptosis to prevent cell proliferation in ovarian cancer. Human ovarian cancer cells and normal human ovarian epithelial cell line IOSE-80 were cultured and administrated with deferoxamine (DFO) or ferric ammonium citrate (FAC). GPX4 knockdown was established for investigating the functions of GPX4 in ovarian cancer cells and in tumor xenograft mice. RESULTS A positively correlation was showed among the levels of GPX4, iron and cell proliferation. Chelation of intracellular iron by DFO disrupted intracellular iron level and was detrimental to ovarian cancer cell survival. FAC-induced elevation of intracellular iron inhibited proliferation, aggravated apoptosis, boosted inflammation and suppressed lipid peroxide reducibility in ovarian cancer cells. Knockdown of GPX4 had similar effects with FAC in ovarian cancer cells. Inhibition of GPX4 suppressed tumor growth, induced ferroptosis, accelerated cell apoptosis, reduced Fe3+ accumulation and suppressed lipid peroxide reducibility in tumor bearing mice. CONCLUSION We demonstrate the significance of GPX4 and intracellular iron level in ovarian cancer cells. Importantly, inhibition of GPX4 interferes with both intracellular iron homeostasis and lipid peroxide reducibility, inducing ferroptosis and exerting anti-cancer effect, which can be a potential effective strategy for ovarian cancer therapy.
Collapse
Affiliation(s)
- Dingxi Li
- Department of Gynecology, Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou City, 450000, Henan Province, China
| | - Mengli Zhang
- Department of Gynecology, Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou City, 450000, Henan Province, China
| | - Hongtu Chao
- Department of Gynecology, Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou City, 450000, Henan Province, China.
| |
Collapse
|
22
|
Ly TTG, Yun J, Lee DH, Chung JS, Kwon SM. Protective Effects and Benefits of Olive Oil and Its Extracts on Women's Health. Nutrients 2021; 13:4279. [PMID: 34959830 PMCID: PMC8705829 DOI: 10.3390/nu13124279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 01/05/2023] Open
Abstract
Women and men share similar diseases; however, women have unique issues, including gynecologic diseases and diseases related to menstruation, menopause, and post menopause. In recent decades, scientists paid more attention to natural products and their derivatives because of their good tolerability and effectiveness in disease prevention and treatment. Olive oil is an essential component in the Mediterranean diet, a diet well known for its protective impact on human well-being. Investigation of the active components in olive oil, such as oleuropein and hydroxytyrosol, showed positive effects in various diseases. Their effects have been clarified in many suggested mechanisms and have shown promising results in animal and human studies, especially in breast cancer, ovarian cancer, postmenopausal osteoporosis, and other disorders. This review summarizes the current evidence of the role of olives and olive polyphenols in women's health issues and their potential implications in the treatment and prevention of health problems in women.
Collapse
Affiliation(s)
- Thanh Truong Giang Ly
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Jisoo Yun
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Dong-Hyung Lee
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea;
| | - Joo-Seop Chung
- Department of Hematology-Oncology, Medical Research Institute, Pusan National University Hospital, Busan 49241, Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; (T.T.G.L.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
23
|
Liu Y, Zhu K, Guan X, Xie S, Wang Y, Tong Y, Guo L, Zheng H, Lu R. TTK is a potential therapeutic target for cisplatin-resistant ovarian cancer. J Ovarian Res 2021; 14:128. [PMID: 34598710 PMCID: PMC8487155 DOI: 10.1186/s13048-021-00884-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/19/2021] [Indexed: 12/17/2022] Open
Abstract
Background Drug resistance and recurrence are main contributors to the poor prognosis of ovarian cancer. Cisplatin is a platinum compound which is widely used in the treatment of various solid tumors including ovarian cancer. Up to now, the mechanism of cisplatin resistance in ovarian cancer is unclear. Threonine and tyrosine kinase (TTK), an integral part of the spindle assembly checkpoint, may be a potential new target associated with chemotherapy sensitivity. Results TTK was up-regulated in the cisplatin-resistant ovarian cancer cell line. Down-regulation of TTK could recover the sensitivity of cisplatin-resistant ovarian cancer cells to cisplatin treatment. Mechanistically, the PI3K/AKT signaling pathway was activated in cisplatin-resistant cells, and this pathway would be affected by TTK expression. Furthermore, TTK was highly expressed in the tissues of ovarian cancer patients, especially those acquired resistance to cisplatin. Conclusions Our study revealed that TTK may be a promising therapeutic target for cisplatin-resistant ovarian cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-021-00884-z.
Collapse
Affiliation(s)
- Yixuan Liu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Keyu Zhu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaolin Guan
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Suhong Xie
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China
| | - Yanchun Wang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China
| | - Ying Tong
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Zheng
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China.
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No.270, Dong'An Road, Xuhui District, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Dysfunctional activity of classical DNA end-joining renders acquired resistance to carboplatin in human ovarian cancer cells. Cancer Lett 2021; 520:267-280. [PMID: 34375710 DOI: 10.1016/j.canlet.2021.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/13/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the deadliest gynecological malignancy worldwide. Although chemotherapy is required as the most standard treatment strategy for ovarian cancer, the survival rates are very low, largely because of high incidence of recurrence due to resistance to conventional surgery and genotoxic chemotherapies. Carboplatin-resistant ovarian cancer cells were generated by continuous treatment over six months. Carboplatin-resistance induced morphological alterations and promoted the rates of proliferation and migration of SKOV3 compared to the parental cells. Interestingly, carboplatin-resistant SKOV3 showed the high levels of γH2AX foci formed at the basal level, and the levels of γH2AX foci remained even after the recovery time, suggesting that the DNA damage response and repair machinery were severely attenuated by carboplatin-resistance. Surprisingly, the expression levels of XRCC4, a critical factor in non-homologous end joining (NHEJ) DNA repair, were significantly decreased in carboplatin-resistant SKOV3 compared with those in non-resistant controls. Furthermore, restoration of NHEJ in carboplatin-resistant SKOV3 by suppression of ABCB1 and/or AR re-sensitized carboplatin-resistant cells to genotoxic stress and reduced their proliferation ability. Our findings suggest that attenuation of the NHEJ DNA repair machinery mediated by resistance to genotoxic stress might be a critical cause of chemoresistance in patients with ovarian cancer.
Collapse
|
25
|
Eckert MA, Orozco C, Xiao J, Javellana M, Lengyel E. The Effects of Chemotherapeutics on the Ovarian Cancer Microenvironment. Cancers (Basel) 2021; 13:3136. [PMID: 34201616 PMCID: PMC8268261 DOI: 10.3390/cancers13133136] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 12/31/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is characterized by a complex and dynamic tumor microenvironment (TME) composed of cancer-associated fibroblasts (CAFs), immune cells, endothelial cells, and adipocytes. Although most approved therapies target cancer cells, a growing body of evidence suggests that chemotherapeutic agents have an important role in regulating the biology of the diverse cells that compose the TME. Understanding how non-transformed cells respond and adapt to established therapeutics is necessary to completely comprehend their action and develop novel therapeutics that interrupt undesired tumor-stroma interactions. Here, we review the effects of chemotherapeutic agents on normal cellular components of the host-derived TME focusing on CAFs. We concentrate on therapies used in the treatment of HGSOC and synthesize findings from studies focusing on other cancer types and benign tissues. Agents such as platinum derivatives, taxanes, and PARP inhibitors broadly affect the TME and promote or inhibit the pro-tumorigenic roles of CAFs by modifying the bidirectional cross-talk between tumor and stromal cells in the tumor organ. While most chemotherapy research focuses on cancer cells, these studies emphasize the need to consider all cell types within the tumor organ when evaluating chemotherapeutics.
Collapse
Affiliation(s)
| | | | | | | | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (M.A.E.); (C.O.); (J.X.); (M.J.)
| |
Collapse
|
26
|
Casper AC, Parsons MW, Chipman J, Burt LM, Suneja G, Maurer KA, Gaffney DK. Risk of secondary malignancies in ovarian cancer survivors: 52,680 patients analyzed with over 40 years of follow-up. Gynecol Oncol 2021; 162:454-460. [PMID: 34092413 DOI: 10.1016/j.ygyno.2021.05.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Survivors of ovarian cancer are at risk of developing a secondary malignancy (SM). We sought to evaluate the risk of developing SM, stratified by treatment modality. METHODS Standardized incidence ratios (SIR, observed-to-expected [O/E] ratio) were assessed in 52,680 patients diagnosed with ovarian cancer between 1975 and 2016 in the National Cancer Institute's Surveillance, Epidemiology, and End Results Program. RESULTS Of the 52,680 patients, 3366 patients (6.4%) developed SM, which was more than the endemic rate (O/E 1.13; p < .05). Patients who received any radiation (RT) had an increased risk of overall SM compared to those who didn't (O/E 1.42 vs 1.11; p < .05), and specifically, in the bladder (O/E 2.81). Patients who received any chemotherapy (CT) had an increased risk of leukemia (O/E 3.06), and a similar risk of overall SM compared to those not treated with CT (O/E 1.11 vs 1.14; p < .05). The excess risk of developing a solid tumor SM was greatest at latencies of 10-20 years. Patients younger than 50 had the highest risk of developing SM. Non-White patients had a higher risk of SM compared to White patients. CONCLUSIONS This is the largest study to examine the risk of SM in patients with ovarian cancer and has the longest follow-up. Risk of SM was increased after ovarian cancer and varied with treatment modality, race, latency, and age. These results may help inform SM screening protocols for women diagnosed with ovarian cancer.
Collapse
Affiliation(s)
- Anthony C Casper
- Rocky Vista University College of Osteopathic Medicine, 255 E Center St, Ivins, UT 84738, USA; Department of Radiation Oncology, Huntsman Cancer Hospital, University of Utah, 2000 Circle of Hope Drive #1950, Salt Lake City, UT 84112, USA.
| | - Matthew W Parsons
- Department of Radiation Oncology, Huntsman Cancer Hospital, University of Utah, 2000 Circle of Hope Drive #1950, Salt Lake City, UT 84112, USA.
| | - Jonathan Chipman
- University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive #1950, Salt Lake City, UT 84112, USA.
| | - Lindsay M Burt
- Department of Radiation Oncology, Huntsman Cancer Hospital, University of Utah, 2000 Circle of Hope Drive #1950, Salt Lake City, UT 84112, USA.
| | - Gita Suneja
- Department of Radiation Oncology, Huntsman Cancer Hospital, University of Utah, 2000 Circle of Hope Drive #1950, Salt Lake City, UT 84112, USA.
| | - Kathryn A Maurer
- University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive #1950, Salt Lake City, UT 84112, USA.
| | - David K Gaffney
- Department of Radiation Oncology, Huntsman Cancer Hospital, University of Utah, 2000 Circle of Hope Drive #1950, Salt Lake City, UT 84112, USA.
| |
Collapse
|
27
|
Zhu M, Ye C, Wang J, Yang G, Ying X. Activation of COL11A1 by PRRX1 promotes tumor progression and radioresistance in ovarian cancer. Int J Radiat Biol 2021; 97:958-967. [PMID: 33970764 DOI: 10.1080/09553002.2021.1928780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/19/2021] [Accepted: 05/03/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Although radiotherapy is a common treatment option for all kinds of cancer patients, including ovarian cancer, a major obstacle limiting its application in the development of resistance. Therefore, it is urgently needed to clarify the mechanism of radiosensitivity modulation. MATERIALS AND METHODS We obtained open datasets and analyzed the expression of collagen type XI alpha 1 (COL11A1) in ovarian cancer patients with different stages. Meanwhile, the correlation of COL11A1 and survival outcomes is determined by Kaplan-Meier analysis. The role of COL11A1 in cell proliferation was observed in an in vitro knockdown system. SKOV3 radioresistant cells were established to determine the role of COL11A1 on radioresistant in ovarian cancer. RESULTS AND DISCUSSION COL11A1 were highly enriched in late-stage ovarian cancer tumor tissues and negatively correlated with survival outcomes in ovarian cancer. The functional analysis found that COL11A1 promoted ovarian cancer cell proliferation in vitro. Importantly, COL11A1 decreased radiosensitivity in ovarian cancer by AKT activation. Paired related homeobox 1 (PRRX1) acted as an upstream transcription factor to regulate COL11A1 expression in ovarian cancer. Increased COL11A1 expression is related to low survival outcomes and radiosensitivity in ovarian cancer. CONCLUSIONS Targeting COL11A1 is a promising strategy for improving radiotherapy efficiency.
Collapse
Affiliation(s)
- Miaomiao Zhu
- Department of Obstetrics and Gynecology, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenxia Ye
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Wang
- Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Guangxia Yang
- Department of Rheumatology, the Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoyan Ying
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
28
|
Liu J, Wu Q, Wu S, Xie X. Investigation on potential biomarkers of hyperprogressive disease (HPD) triggered by immune checkpoint inhibitors (ICIs). Clin Transl Oncol 2021; 23:1782-1793. [PMID: 33847923 DOI: 10.1007/s12094-021-02579-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/01/2021] [Indexed: 02/04/2023]
Abstract
PURPOSE This project aimed to survey the clinical characteristics and survivals of hyperprogressive disease (HPD) mediated by immune checkpoint inhibitors (ICIs) in an attempt to explore the potential predictors. METHODS After searching PubMed, MEDLINE, Google Scholar and Cochrane Library databases, 12 studies incorporating 1766 individuals were enrolled. The data were analyzed with Review manager 5.3 software. RESULTS The results revealed HPD correlated with previous metastatic sites > 2 (OR = 1.86, 95% CI 1.33-2.59, P = 0.0003), liver metastasis (OR = 3.35, 95% CI 2.09-5.35, P < 0.00001), Royal Marsden Hospital (RMH) score ≥ 2 (OR = 2.80, 95% CI 1.85-4.23, P < 0.00001), higher ECOG PS (OR = 1.60, 95% CI 1.13-2.27, P = 0.008) and LDH > upper limits of normal (ULN) (OR = 2.32, 95% CI 1.51-3.58, P = 0.0001). Instead, HPD was unrelated to gender, age, smoking status, PD-L1 expression, therapy, neutrophil-to-lymphocyte ratio, the histology, the status of EGFR, ALK and KRAS in non-small cell lung cancer and HER-2 expression in advanced gastric cancer. Moreover, HPD was evidently correlated with a shorter OS (HR = 2.92, 95% CI 1.79-4.76, P < 0.0001) and PFS (HR = 3.62, 95% CI 2.79-4.68, P < 0.00001). The same phenomena existed in stratified studies based on study regions and tumor types. CONCLUSIONS This study demonstrated that HPD was related to the number of prior metastatic sites > 2, liver metastasis, RMH score ≥ 2, higher ECOG PS score and LDH > ULN. Moreover, HPD was correlated with a poor OS and PFS in patients following ICI therapy.
Collapse
Affiliation(s)
- J Liu
- Department of Oncology, Molecular Oncology Research Institute, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China
| | - Q Wu
- Department of Oncology, Molecular Oncology Research Institute, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China
| | - S Wu
- Department of Oncology, Molecular Oncology Research Institute, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China
| | - X Xie
- Department of Oncology, Molecular Oncology Research Institute, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China. .,Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China.
| |
Collapse
|
29
|
Stevens MJ, West S, Gard G, Renaud C, Nevell D, Roderick S, Le A. Utility of adjuvant whole abdominal radiation therapy in ovarian clear cell cancer (OCCC): a pragmatic cohort study of women with classic immuno-phenotypic signature. Radiat Oncol 2021; 16:29. [PMID: 33549120 PMCID: PMC7866446 DOI: 10.1186/s13014-021-01750-4] [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: 05/07/2020] [Accepted: 01/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To evaluate the initial experience and clinical utility of first-line adjuvant intensity-modulated whole abdominal radiation therapy (WART) in women with ovarian clear cell cancer (OCCC) referred to an academic center. METHODS Progression-free and overall survival was analyzed in a pragmatic observational cohort study of histologically pure OCCC patients over-expressing HNF-1ß treated between 2013 and end-December 2018. An in-house intensity-modulated WART program was developed from a published pre-clinical model. Radiation dose-volume data was curated to American Association of Physics in Medicine (AAPM) Task Group 263 recommendations. A dedicated database prospectively recorded presenting characteristics and outcomes in a standardized fashion. RESULTS Five women with FIGO (2018) stage IA to IIIA2 OCCC were treated with first-line WART. Median age was 58 years (range 47-68 years). At diagnosis CA-125 was elevated in 4 cases (median 56 kU/L: range 18.4-370 kU/L) before primary de-bulking surgery. Severe premorbid endometriosis was documented in 3 patients. At a median follow-up of 77 months (range 16-83 mo.), all patients remain alive and progression-free on clinical, biochemical (CA-125), and 18Fluoro-deoxyglucose (FDG) PET/CT re-evaluation. Late radiation toxicity was significant (G3) in 1 case who required a limited bowel resection and chronic nutritional support at 9 months post-WART; 2 further patients had asymptomatic (G2) osteoporotic fragility fractures of axial skeleton at 12 months post-radiation treated with anti-resorptive agents (denosumab). CONCLUSIONS The clinical utility of intensity-modulated WART in OCCC over-expressing HNF-1β was suggested in this small observational cohort study. The hypothesis that HNF-1β is a portent of platinum-resistance and an important predictive biomarker in OCCC needs further confirmation. Curating multi-institutional cohort studies utilizing WART by means of "Big Data" may improve OCCC care standards in the future.
Collapse
Affiliation(s)
- Mark J Stevens
- Department of Radiation Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Level 1 ASB Building, St Leonards, NSW, 2065, Australia. .,Northern Clinical School, University of Sydney, St Leonards, NSW, Australia.
| | - Simon West
- Northern Clinical School, University of Sydney, St Leonards, NSW, Australia.,Department of Obstetrics and Gynecology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Gregory Gard
- Northern Clinical School, University of Sydney, St Leonards, NSW, Australia.,Department of Obstetrics and Gynecology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Christopher Renaud
- Department of Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - David Nevell
- Department of Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Stephanie Roderick
- Department of Radiation Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Level 1 ASB Building, St Leonards, NSW, 2065, Australia
| | - Andrew Le
- Department of Radiation Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Level 1 ASB Building, St Leonards, NSW, 2065, Australia
| |
Collapse
|
30
|
Angelicola S, Ruzzi F, Landuzzi L, Scalambra L, Gelsomino F, Ardizzoni A, Nanni P, Lollini PL, Palladini A. IFN-γ and CD38 in Hyperprogressive Cancer Development. Cancers (Basel) 2021; 13:309. [PMID: 33467713 PMCID: PMC7830527 DOI: 10.3390/cancers13020309] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) improve the survival of patients with multiple types of cancer. However, low response rates and atypical responses limit their success in clinical applications. The paradoxical acceleration of tumor growth after treatment, defined as hyperprogressive disease (HPD), is the most difficult problem facing clinicians and patients alike. The mechanisms that underlie hyperprogression (HP) are still unclear and controversial, although different factors are associated with the phenomenon. In this review, we propose two factors that have not yet been demonstrated to be directly associated with HP, but upon which it is important to focus attention. IFN-γ is a key cytokine in antitumor response and its levels increase during ICI therapy, whereas CD38 is an alternative immune checkpoint that is involved in immunosuppressive responses. As both factors are associated with resistance to ICI therapy, we have discussed their possible involvement in HPD with the conclusion that IFN-γ may contribute to HP onset through the activation of the inflammasome pathway, immunosuppressive enzyme IDO1 and activation-induced cell death (AICD) in effector T cells, while the role of CD38 in HP may be associated with the activation of adenosine receptors, hypoxia pathways and AICD-dependent T-cell depletion.
Collapse
Affiliation(s)
- Stefania Angelicola
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Lorena Landuzzi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Laura Scalambra
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Francesco Gelsomino
- Divisione di Oncologia Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.G.); (A.A.)
| | - Andrea Ardizzoni
- Divisione di Oncologia Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.G.); (A.A.)
| | - Patrizia Nanni
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Arianna Palladini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| |
Collapse
|
31
|
Butala AA, Patel RR, Manjunath S, Latif NA, Haggerty AF, Jones JA, Taunk NK. Palliative Radiation Therapy for Metastatic, Persistent, or Recurrent Epithelial Ovarian Cancer: Efficacy in the Era of Modern Technology and Targeted Agents. Adv Radiat Oncol 2021; 6:100624. [PMID: 33665491 PMCID: PMC7897761 DOI: 10.1016/j.adro.2020.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/10/2020] [Accepted: 11/11/2020] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Metastatic, persistent, or recurrent epithelial ovarian cancer (MPR-EOC) remains a significant threat to patient mortality despite advances in novel targeted agents. Radiation therapy (RT) is often used as a palliative option. We report outcomes of a large series of MPR-EOC patients treated with modern palliative RT (PRT) in an era of novel systemic therapies. METHODS AND MATERIALS A retrospective review was conducted of women treated with PRT for MPR-EOC between 2007 and 2019 at an academic institution. Clinical response rates were recorded at <1 month, 1 to 3 months, and >3 months. Radiographic responses were categorized by RECIST 1.1 criteria. Overall response rate (ORR) was the sum of complete and partial response. Linear regression analyses of baseline characteristics were conducted for statistical testing. RESULTS Eighty-six patients with PMR-OC received 120 courses of palliative RT. Median follow-up was 8.6 months. Median age was 61 (range, 22-82). Thirty-six percent of women received central nervous system (CNS)-directed RT. In addition, 43% received targeted therapies before RT. Clinical ORR within 1 month and at last follow-up for non-CNS lesions was 79% and 61% (69% and 88% for CNS lesions, respectively). High-grade serous lesions were more likely to have clinical response (P = .04). Biologically effective doses (BED) >39 Gy were associated with improved clinical response in CNS lesions (P = .049). Bony sites were associated with worse clinical (P = .004) response in non-CNS lesions compared with soft tissue or nodal sites. Acute or late grade 3+ toxicities with bevacizumab were low (8.7%/4.3%). CONCLUSIONS PRT offers excellent rates of response for symptomatic patients with MPR-EOC within 1 month of treatment, with durable responses beyond 3 months. High-grade serous lesions were associated with improved response in all patients. Higher BED and soft tissue or nodal sites were associated with improved response in CNS and non-CNS patients, respectively. Acute or late toxicities with bevacizumab and PRT were low. Prospective investigation is warranted to determine the optimal PRT regimen.
Collapse
Affiliation(s)
- Anish A. Butala
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Shwetha Manjunath
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nawar A. Latif
- Department of Gynecologic Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ashley F. Haggerty
- Department of Gynecologic Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua A. Jones
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Neil K. Taunk
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
32
|
Jin ZH, Tsuji AB, Degardin M, Sugyo A, Obara S, Wakizaka H, Nagatsu K, Hu K, Zhang MR, Dumy P, Boturyn D, Higashi T. Radiotheranostic Agent 64Cu-cyclam-RAFT-c(-RGDfK-) 4 for Management of Peritoneal Metastasis in Ovarian Cancer. Clin Cancer Res 2020; 26:6230-6241. [PMID: 32933998 DOI: 10.1158/1078-0432.ccr-20-1205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/11/2020] [Accepted: 09/10/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Ovarian cancer peritoneal metastases (OCPMs) are a pathophysiologically heterogeneous group of tumors that are rarely curable. αVβ3 integrin (αVβ3) is overexpressed on tumoral neovessels and frequently on ovarian cancer cells. Here, using two clinically relevant αVβ3-positive OCPM mouse models, we studied the theranostic potential of an αVβ3-specific radiopeptide, 64Cu-cyclam-RAFT-c(-RGDfK-)4 (64Cu-RaftRGD), and its intra- and intertumoral distribution in relation to the tumor microenvironment. EXPERIMENTAL DESIGN αVβ3-expressing peritoneal and subcutaneous models of ovarian carcinoma (IGR-OV1 and NIH:OVCAR-3) were established in nude mice. 64Cu-RaftRGD was administered either intravenously or intraperitoneally. We performed intratumoral distribution (ITD) studies, PET/CT imaging and quantification, biodistribution assay and radiation dosimetry, and therapeutic efficacy and toxicity studies. RESULTS Intraperitoneal administration was an efficient route for targeting 64Cu-RaftRGD to OCPMs with excellent tumor penetration. Using the fluorescence surrogate, Cy5.5-RaftRGD, in our unique high-resolution multifluorescence analysis, we found that the ITD of 64Cu-RaftRGD was spatially distinct from, but complementary to, that of hypoxia. 64Cu-RaftRGD-based PET enabled clear visualization of multiple OCPM deposits and ascites and biodistribution analysis demonstrated an inverse correlation between tumor uptake and tumor size (1.2-17.2 mm). 64Cu-RaftRGD at a radiotherapeutic dose (148 MBq/0.357 nmol) showed antitumor activities by inhibiting tumor cell proliferation and inducing apoptosis, with negligible toxicity. CONCLUSIONS Collectively, these results demonstrate the all-in-one potential of 64Cu-RaftRGD for imaging guided radiotherapy of OCPM by targeting both tumoral neovessels and cancerous cells. On the basis of the ITD finding, we propose that pairing αVβ3- and hypoxia-targeted radiotherapies could improve therapeutic efficacy by overcoming the heterogeneity of ITD encountered with single-agent treatments.
Collapse
Affiliation(s)
- Zhao-Hui Jin
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| | - Atsushi B Tsuji
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| | | | - Aya Sugyo
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Satoshi Obara
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hidekatsu Wakizaka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kotaro Nagatsu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kuan Hu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Pascal Dumy
- Institut des Biomolécules Max Mousseron, École Nationale Supérieure de Chimie de Montpellier, Université de Montpellier, Montpellier, France
| | | | - Tatsuya Higashi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
33
|
Norell CH, Butler J, Farrell R, Altman A, Bentley J, Cabasag CJ, Cohen PA, Fegan S, Fung-Kee-Fung M, Gourley C, Hacker NF, Hanna L, Høgdall CK, Kristensen G, Kwon J, McNally O, Nelson G, Nordin A, O'Donnell D, Schnack T, Sykes PH, Zotow E, Harrison S. Exploring international differences in ovarian cancer treatment: a comparison of clinical practice guidelines and patterns of care. Int J Gynecol Cancer 2020; 30:1748-1756. [PMID: 32784203 PMCID: PMC7656152 DOI: 10.1136/ijgc-2020-001403] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION The International Cancer Benchmarking Partnership demonstrated international differences in ovarian cancer survival, particularly for women aged 65-74 with advanced disease. These findings suggest differences in treatment could be contributing to survival disparities. OBJECTIVE To compare clinical practice guidelines and patterns of care across seven high-income countries. METHODS A comparison of guidelines was performed and validated by a clinical working group. To explore clinical practice, a patterns of care survey was developed. A questionnaire regarding management and potential health system-related barriers to providing treatment was emailed to gynecological specialists. Guideline and survey results were crudely compared with 3-year survival by 'distant' stage using Spearman's rho. RESULTS Twenty-seven guidelines were compared, and 119 clinicians completed the survey. Guideline-related measures varied between countries but did not correlate with survival internationally. Guidelines were consistent for surgical recommendations of either primary debulking surgery or neoadjuvant chemotherapy followed by interval debulking surgery with the aim of complete cytoreduction. Reported patterns of surgical care varied internationally, including for rates of primary versus interval debulking, extensive/'ultra-radical' surgery, and perceived barriers to optimal cytoreduction. Comparison showed that willingness to undertake extensive surgery correlated with survival across countries (rs=0.94, p=0.017). For systemic/radiation therapies, guideline differences were more pronounced, particularly for bevacizumab and PARP (poly (ADP-ribose) polymerase) inhibitors. Reported health system-related barriers also varied internationally and included a lack of adequate hospital staffing and treatment monitoring via local and national audits. DISCUSSION Findings suggest international variations in ovarian cancer treatment. Characteristics relating to countries with higher stage-specific survival included higher reported rates of primary surgery; willingness to undertake extensive/ultra-radical procedures; greater access to high-cost drugs; and auditing.
Collapse
Affiliation(s)
- Charles H Norell
- International Cancer Benchmarking Partnership (ICBP), Policy & Information, Cancer Research UK, London, UK
| | - John Butler
- International Cancer Benchmarking Partnership (ICBP), Policy & Information, Cancer Research UK, London, UK
- Gynaecology Department, Royal Marsden NHS Foundation Trust, London, UK
| | - Rhonda Farrell
- Gynaecological Oncology Department, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Alon Altman
- Department of Gynecologic Oncology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James Bentley
- Department of Obstetrics and Gynaecology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Citadel J Cabasag
- Section of Cancer Surveillance, International Agency for Research on Cancer, Lyon, France
| | - Paul A Cohen
- Department of Gynaecological Oncology, St John of God Health Care, West Perth, Ontario, Australia
| | - Scott Fegan
- Department of Obstetrics and Gynaecology, NHS Lothian, Edinburgh, UK
| | - Michael Fung-Kee-Fung
- Department of Obstetrics, Gynaecology and Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Charlie Gourley
- Cancer Research UK Edinburgh Centre, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Neville F Hacker
- Gynaecological Cancer Centre, Royal Hospital for Women, Randwick, New South Wales, Australia
- School of Women's & Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Louise Hanna
- Department of Oncology, Velindre Cancer Centre, Cardiff, Wales, UK
| | - Claus Kim Høgdall
- Gynecologic Department, The Juliane Marie Centre, Copenhagen, Denmark
| | - Gunnar Kristensen
- Department of Gynecologic Oncology and Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Janice Kwon
- Department of Obstetrics and Gynaecology, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Orla McNally
- Oncology and Dysplasia Service, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Gregg Nelson
- Department of Oncology, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Andy Nordin
- Department of Gynaecological Oncology, East Kent Hospitals University NHS Foundation Trust, Canterbury, UK
| | | | - Tine Schnack
- Department of Gynaecology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Peter H Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Dunedin, New Zealand
| | - Ewa Zotow
- Policy & Information, Cancer Research UK, London, UK
| | | |
Collapse
|
34
|
Horowitz M, Esakov E, Rose P, Reizes O. Signaling within the epithelial ovarian cancer tumor microenvironment: the challenge of tumor heterogeneity. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:905. [PMID: 32793749 DOI: 10.21037/atm-2019-cm-08] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epithelial ovarian cancer (EOC) is a leading cause of cancer death in women. Standard of care treatment has remained platinum-containing cytotoxic chemotherapy for over three decades. Among the central challenges in treating ovarian CA are disease recurrence and the development of chemoresistance. Survival is uniformly poor for patients with chemoresistant recurrent disease and effective therapeutic options are limited. As such, delineating the mechanisms of chemoresistance and developing targeted therapies to prevent chemoresistance from occurring are of vital importance to improving survival for patients with EOC. Attempts to characterize mechanisms of chemoresistance have implicated numerous cellular pathways, but a rift remains between pre-clinical findings and translation to improving patient survival. More recently, the interplay among different cell types within the tumor microenvironment has become central to understanding how chemoresistance may develop and may be sustained. An improved understanding of how tumor cell-intrinsic and -extrinsic pathways converge during the development of chemoresistance may improve the likelihood of successful clinical translation. This review focuses on the roles of the EOC tumor microenvironment and tumor cell heterogeneity in the development of chemoresistance. We review recent studies into mechanisms of chemoresistance as they relate to tumor microenvironment and development of novel therapeutic approaches that exploit these mechanisms to prevent or reverse chemoresistance. This review attempts to cast these latest discoveries in a clinical context by summarizing trends in ongoing clinical trials for patients with EOC.
Collapse
Affiliation(s)
- Max Horowitz
- Division of Gynecologic Oncology, Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Obstetrics & Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Emily Esakov
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Peter Rose
- Division of Gynecologic Oncology, Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Obstetrics & Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ofer Reizes
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
35
|
DNA Repair and Ovarian Carcinogenesis: Impact on Risk, Prognosis and Therapy Outcome. Cancers (Basel) 2020; 12:cancers12071713. [PMID: 32605254 PMCID: PMC7408288 DOI: 10.3390/cancers12071713] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022] Open
Abstract
There is ample evidence for the essential involvement of DNA repair and DNA damage response in the onset of solid malignancies, including ovarian cancer. Indeed, high-penetrance germline mutations in DNA repair genes are important players in familial cancers: BRCA1, BRCA2 mutations or mismatch repair, and polymerase deficiency in colorectal, breast, and ovarian cancers. Recently, some molecular hallmarks (e.g., TP53, KRAS, BRAF, RAD51C/D or PTEN mutations) of ovarian carcinomas were identified. The manuscript overviews the role of DNA repair machinery in ovarian cancer, its risk, prognosis, and therapy outcome. We have attempted to expose molecular hallmarks of ovarian cancer with a focus on DNA repair system and scrutinized genetic, epigenetic, functional, and protein alterations in individual DNA repair pathways (homologous recombination, non-homologous end-joining, DNA mismatch repair, base- and nucleotide-excision repair, and direct repair). We suggest that lack of knowledge particularly in non-homologous end joining repair pathway and the interplay between DNA repair pathways needs to be confronted. The most important genes of the DNA repair system are emphasized and their targeting in ovarian cancer will deserve further attention. The function of those genes, as well as the functional status of the entire DNA repair pathways, should be investigated in detail in the near future.
Collapse
|
36
|
Chuang YC, Chu CH, Cheng SH, Liao LD, Chu TS, Chen NT, Paldino A, Hsia Y, Chen CT, Lo LW. Annealing-modulated nanoscintillators for nonconventional X-ray activation of comprehensive photodynamic effects in deep cancer theranostics. Theranostics 2020; 10:6758-6773. [PMID: 32550902 PMCID: PMC7295068 DOI: 10.7150/thno.41752] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 05/04/2020] [Indexed: 01/10/2023] Open
Abstract
Photodynamic therapy (PDT), which involves the generation of reactive oxygen species (ROS) through interactions of a photosensitizer (PS) with light and oxygen, has been applied in oncology. Over the years, PDT techniques have been developed for the treatment of deep-seated cancers. However, (1) the tissue penetration limitation of excitation photon, (2) suppressed efficiency of PS due to multiple energy transfers, and (3) insufficient oxygen source in hypoxic tumor microenvironment still constitute major challenges facing the clinical application of PDT for achieving effective treatment. We present herein a PS-independent, ionizing radiation-induced PDT agent composed of yttrium oxide nanoscintillators core and silica shell (Y2O3:Eu@SiO2) with an annealing process. Our results revealed that annealed Y2O3:Eu@SiO2 could directly induce comprehensive photodynamic effects under X-ray irradiation without the presence of PS molecules. The crystallinity of Y2O3:Eu@SiO2 was demonstrated to enable the generation of electron-hole (e--h+) pairs in Y2O3 under ionizing irradiation, giving rise to the formation of ROS including superoxide, hydroxyl radical and singlet oxygen. In particular, combining Y2O3:Eu@SiO2 with fractionated radiation therapy increased radio-resistant tumor cell damage. Furthermore, photoacoustic imaging of tumors showed re-distribution of oxygen saturation (SO2) and reoxygenation of the hypoxia region. The results of this study support applicability of the integration of fractionated radiation therapy with Y2O3:Eu@SiO2, achieving synchronously in-depth and oxygen-insensitive X-ray PDT. Furthermore, we demonstrate Y2O3:Eu@SiO2 exhibited radioluminescence (RL) under X-ray irradiation and observed the virtually linear correlation between X-ray-induced radioluminescence (X-RL) and the Y2O3:Eu@SiO2 concentration in vivo. With the pronounced X-RL for in-vivo imaging and dosimetry, it possesses significant potential for utilization as a precision theranostics producing highly efficient X-ray PDT for deep-seated tumors.
Collapse
|
37
|
Kowalchuk RO, Waters MR, Richardson KM, Spencer K, Larner JM, Irvin WP, Kersh CR. Stereotactic body radiation therapy in the treatment of ovarian cancer. Radiat Oncol 2020; 15:108. [PMID: 32404167 PMCID: PMC7222303 DOI: 10.1186/s13014-020-01564-w] [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: 03/03/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study evaluates the outcomes and toxicity of stereotactic body radiation therapy (SBRT) in ovarian cancer. METHODS This retrospective analysis considered all patients treated with SBRT from 2009 to 2018 with a primary ovarian tumor. Follow-up included PET-CT and CT scans at 2-3 month intervals. Statistical analysis primarily consisted of univariate analysis, Cox proportional hazards analysis, and the Kaplan-Meier method. RESULTS The study included 35 patients with 98 treatments for lymph nodes (51), local recurrence (21), and de novo solid metastases (26). Median biologically effective dose (BED), gross tumor volume, and planning target volume were 38.40 Gy, 10.41 cc, and 25.21 cc, respectively. 52 lesions showed complete radiographic response, and two-year local control was 80%. Median overall survival (OS) was 35.2 months, and two-year progression-free survival (PFS) was 12%. On univariate analysis, Eastern Cooperative Oncology Group performance status > 0 was predictive of decreased OS (p = 0.0024) and PFS (p = 0.044). Factors predictive of local failure included lower BED (p = 0.016), treatment for recurrence (p = 0.029), and higher pre-treatment SUV (p = 0.026). Kaplan-Meier analysis showed BED ≤35 Gy (p < 0.005) and treatment for recurrence (p = 0.01) to be predictive of local failure. On Cox proportional hazards analysis, treatment of lymph nodes was predictive of complete radiographic response (hazard ratio (HR) = 4.95), as was higher BED (HR = 1.03). Toxicity included 27 cases of grade < 3 toxicity, and one grade 5 late toxicity of GI bleed from a radiation therapy-induced duodenal ulcer. CONCLUSIONS SBRT provides durable local control with minimal toxicity in ovarian cancer, especially with BED > 35 Gy and treatment for lymph nodes.
Collapse
Affiliation(s)
- Roman O Kowalchuk
- University of Virginia / Riverside, Radiosurgery Center, Newport News, VA, USA.
| | - Michael R Waters
- University of Virginia / Riverside, Radiosurgery Center, Newport News, VA, USA
| | - K Martin Richardson
- University of Virginia / Riverside, Radiosurgery Center, Newport News, VA, USA
| | - Kelly Spencer
- University of Virginia / Riverside, Radiosurgery Center, Newport News, VA, USA
| | - James M Larner
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA, USA
| | - William P Irvin
- Department of Gynecologic Oncology, Riverside Regional Medical Center, Newport News, USA
| | - Charles R Kersh
- University of Virginia / Riverside, Radiosurgery Center, Newport News, VA, USA
| |
Collapse
|
38
|
Abu-Tineh M, Elmalik H, Yassin MA. Metastatic Ovarian Cancer Presenting as Inflammatory Breast Cancer: A Case Report. Case Rep Oncol 2020; 13:867-874. [PMID: 32884533 PMCID: PMC7443636 DOI: 10.1159/000508358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/03/2020] [Indexed: 11/19/2022] Open
Abstract
Metastatic ovarian cancer to the breast is a rare presentation, with limited cases reported worldwide. Common sites for distant metastasis in ovarian cancer are to the liver, lung, and pleura [Dauplat et al. Cancer. 1987 Oct 1;60(7):1561-6]. Usually, such cases predict poor prognosis with troublesome management. We present one challenging case of a 54-year-old female patient with recurrent clear cell ovarian cancer, presenting with right breast mass of primary versus secondary origin, progressing into inflammatory breast cancer picture. Our report aims to shed light on the value of early suspicion and low threshold of detecting secondary breast masses of ovarian cancer origin.
Collapse
Affiliation(s)
| | - Hind Elmalik
- National Center for Cancer Care and Research, Department of Oncology, Hamad Medical Corporation, Doha, Qatar
| | - Mohamed A. Yassin
- National Center for Cancer Care and Research, Department of Oncology, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
39
|
Terraneo N, Jacob F, Dubrovska A, Grünberg J. Novel Therapeutic Strategies for Ovarian Cancer Stem Cells. Front Oncol 2020; 10:319. [PMID: 32257947 PMCID: PMC7090172 DOI: 10.3389/fonc.2020.00319] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is one of the most lethal gynecologic malignancies. Due to the lack of specific symptoms and screening methods, this disease is usually diagnosed only at an advanced and metastatic stage. The gold-standard treatment for OC patients consists of debulking surgery followed by taxane combined with platinum-based chemotherapy. Most patients show complete clinical remission after first-line therapy, but the majority of them ultimately relapse, developing radio- and chemoresistant tumors. It is now proposed that the cause of recurrence and reduced therapy efficacy is the presence of small populations of cancer stem cells (CSCs). These cells are usually resistant against conventional cancer therapies and for this reason, effective targeted therapies for the complete eradication of CSCs are urgently needed. In this review article, we highlight the mechanisms of CSC therapy resistance, epithelial-to-mesenchymal transition, stemness, and novel therapeutic strategies for ovarian CSCs.
Collapse
Affiliation(s)
- Nastassja Terraneo
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
| | - Francis Jacob
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Jürgen Grünberg
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
| |
Collapse
|
40
|
Ellerin BE, Demandante CGN, Martins JT. Pure abscopal effect of radiotherapy in a salivary gland carcinoma: Case report, literature review, and a search for new approaches. Cancer Radiother 2020; 24:226-246. [PMID: 32192840 DOI: 10.1016/j.canrad.2020.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
We report the case of an 84-year-old woman with poorly differentiated non-small cell carcinoma of the right parotid who presented with headache, was found to have a primary right parotid gland cancer as well as metastatic disease, and underwent palliative radiotherapy to the primary site. The patient received no chemotherapy or immunotherapy, but both the primary site and several non-irradiated foci in the lungs regressed or completely resolved. The patient remained free of disease for about one year before progression. The case is a rare instance of abscopal regression of metastatic disease in the absence of pharmacologic immunomodulation. A literature review surveys the history of the abscopal effect of radiation therapy, attempts to understand the mechanisms of its successes and failures, and points to new approaches that can inform and improve the outcomes of radioimmunotherapy.
Collapse
Affiliation(s)
| | | | - J T Martins
- UT Health HOPE Cancer Center, Tyler, TX 75701, USA
| |
Collapse
|
41
|
Immune Checkpoint Inhibitors in Epithelial Ovarian Cancer: An Overview on Efficacy and Future Perspectives. Diagnostics (Basel) 2020; 10:diagnostics10030146. [PMID: 32156035 PMCID: PMC7151145 DOI: 10.3390/diagnostics10030146] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/21/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of death among gynecological cancers. Despite improvements in medical treatments, the prognosis for EOC remains poor, and there is an urgent need for new therapeutic strategies. Immune checkpoint inhibitors (CPIs) have dramatically improved survival of several cancers and are under evaluation in OC. Unfortunately, CPIs have shown globally unsatisfactory results. The aim of this manuscript is to critically review the results from early-phase trials with CPIs in terms of safety and activity, discuss the possible reasons for disappointing results and the new therapeutic approaches to improve patient outcomes.
Collapse
|
42
|
Ma Q, Kai J, Liu Y, Tong Y, Xie S, Zheng H, Wang Y, Guo L, Lu R. Targeting Ku86 enhances X-ray-induced radiotherapy sensitivity in serous ovarian cancer cells. Int J Biochem Cell Biol 2020; 121:105705. [PMID: 32027982 DOI: 10.1016/j.biocel.2020.105705] [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: 10/31/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 11/26/2022]
Abstract
Drug resistance and recurrence are significant contributors to the poor prognosis of serous ovarian cancer (SOC). Radiotherapy is primarily used for the treatment of cancer recurrence; however, it is rarely applied in cases of SOC. Ku86, also known as XRCC5(X-ray repair cross complementing 5), has rarely been reported in the radiotherapy of SOC. Therefore, this study aimed to investigate the role of Ku86 in the development of SOC and in radiotherapy sensitivity induced by X-ray. In vitro experiments and database analysis showed significantly elevated expression of Ku86 in SOC. Further, after down-regulating Ku86 using RNAi technology, cell proliferation was inhibited. Further, the cell cycle was blocked in the G2 phase, and G2/G1 was increased since X-ray irradiation led to an increase in γ-H2AX. Down-regulation of Ku86 in the case of X-ray irradiation will promote the above biological effects, with more γ-H2AX and higher G2/G1. Here, we further deduce that Ku86 promotes the X-ray induced effect is most likely to activate the ATR pathway to block the cell cycle while inhibiting the NHEJ pathway to inhibit DNA damage response(DDR). Together these findings suggest that the down-regulation of Ku86 can improve X-ray-induced radiotherapy by affecting ATR and NHEJ pathways, and provide a new strategy for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Qian Ma
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinyan Kai
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yixuan Liu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Tong
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Suhong Xie
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hui Zheng
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yanchun Wang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
43
|
Leong L, Tan HL, Cua S, Yong KSM, Chen Q, Choo A. Preclinical Activity of Embryonic Annexin A2-Specific Chimeric Antigen Receptor T Cells Against Ovarian Cancer. Int J Mol Sci 2020; 21:ijms21020381. [PMID: 31936170 PMCID: PMC7013580 DOI: 10.3390/ijms21020381] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/30/2019] [Accepted: 01/06/2020] [Indexed: 02/08/2023] Open
Abstract
Chimeric antigen receptors (CARs) have found clinical success in B cell malignancies, but a dearth of potential targets limits their wider clinical application, especially in solid tumours. Here, we describe the development of an anti-annexin A2 CAR, CAR(2448), derived from an antibody found to have activity against epithelial ovarian cancer cell lines. The spacer length of CAR(2448) was optimised based on in vitro cytotoxic activity against ovarian cancer (OC) cell lines via a real-time cytotoxicity assay. The longer spacer CAR(2448)L T cells exhibit significant effector activity, inducing inflammatory cytokine release and cytotoxicity against OC cell lines. Furthermore, CAR(2448)L-BBz T cells induced enhanced survival in an in vivo OC xenograft model and reduced tumour volume by 76.6%. Our preclinical studies of CAR(2448) suggest its potential for the unmet need of novel strategies for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Leonard Leong
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
- NUS Graduate School for Integrative Sciences and Engineering (NGS), National University of Singapore, Singapore 119077, Singapore
| | - Heng Liang Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
| | - Simeon Cua
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
| | - Kylie Su Mei Yong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Andre Choo
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore (NUS), Singapore 117575, Singapore
- Correspondence:
| |
Collapse
|
44
|
Lakomy DS, Urbauer DL, Westin SN, Lin LL. Phase I study of the PARP inhibitor talazoparib with radiation therapy for locally recurrent gynecologic cancers. Clin Transl Radiat Oncol 2019; 21:56-61. [PMID: 31993510 PMCID: PMC6974697 DOI: 10.1016/j.ctro.2019.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/22/2019] [Accepted: 12/26/2019] [Indexed: 12/11/2022] Open
Abstract
PARP inhibitors combined with radiotherapy may be promising for solid tumors. PARP inhibitors radiosensitize ovarian cancer cells both in vitro and in vivo. Talazoparib is a novel PARP-inhibitor with confirmed response in ovarian cancer. We propose a phase I study of talazoparib and concurrent radiation therapy.
PARP inhibitors have been shown to radiosensitize tumor cells in both in vitro and in vivo studies. This is a phase I study that aims to determine the safety, tolerability, and maximally tolerated dose of talazoparib, a PARP inhibitor, when delivered concurrently with radiotherapy in women with recurrent gynecologic cancers.
Collapse
Affiliation(s)
- David S Lakomy
- Dartmouth Geisel School of Medicine, Dartmouth College, Hanover, NH, United States.,Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diana L Urbauer
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shannon N Westin
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lilie L Lin
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
45
|
Chang CYY, Yang PY, Tsai FJ, Li TM, Chiou JS, Chen CJ, Lin TH, Liao CC, Huang SM, Ban B, Liang WM, Lin YJ. Integrated Chinese Herbal Medicine Therapy Improves the Survival of Patients With Ovarian Cancer. Integr Cancer Ther 2019. [PMCID: PMC6902381 DOI: 10.1177/1534735419881497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Ovarian cancer is the seventh most commonly diagnosed
malignancy worldwide and has the highest mortality rate among all gynecological
cancers. Chinese herbal medicine (CHM) is widely applied in Taiwan and has been
used in integrated therapies to treat patients with cancer.
Methods: Patients with ovarian cancer who were registered in
the Taiwan Registry for Catastrophic Illness Patients Database between 1997 and
2012 were considered for this study. A 1:1 individual matching by age was
implemented. A total of 101 CHM users and 101 non-CHM users were involved. A Cox
proportional hazard regression model was applied to evaluate the hazard ratio of
overall mortality. The Kaplan-Meier method and log-rank test were used to
calculate the cumulative incidence of the overall survival rate. Association
rule mining and network analysis were used to analyze CHM prescription patterns.
Results: CHM users showed a significantly lower risk of overall
mortality than nonusers (hazard ratio = 0.45, 95% confidence interval =
0.23-0.91; P = .0256; multivariate Cox proportional hazard
model). The cumulative incidence of the overall survival probability was higher
for CHM users than for non-CHM users (log-rank test, P =
.0009). Association rule mining and network analysis suggested that the main CHM
cluster was associated with the usage of Bu-Zhong-Yi-Qi-Tang, Chuan-Xiong, and
Xi-Xin, followed by the use of Bai-Shao, Da-Huang, and Di-Huang.
Conclusions: CHM, as an adjunctive therapy, may reduce the
overall mortality in patients with ovarian cancer. A list of herbal medicines
that could potentially be used in future studies and clinical trials has also
been provided.
Collapse
Affiliation(s)
| | | | - Fuu-Jen Tsai
- China Medical University Hospital, Taichung
- China Medical University, Taichung
- Asia University, Taichung
| | | | | | - Chao-Jung Chen
- China Medical University Hospital, Taichung
- China Medical University, Taichung
| | | | | | | | - Bo Ban
- Chinese Research Center for Behavior Medicine in Growth and Development, Jining, Shandong, China
| | | | - Ying-Ju Lin
- China Medical University Hospital, Taichung
- China Medical University, Taichung
| |
Collapse
|
46
|
Ordoñez RMLS, Amendola BE, Martinez PF, Wolf A, Coy SR, Amendola M. Radiosurgery for brain metastases from ovarian cancer: an analysis of 25 years' experience with Gamma Knife treatment. Rep Pract Oncol Radiother 2019; 24:667-671. [PMID: 31719805 DOI: 10.1016/j.rpor.2019.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 07/18/2019] [Accepted: 10/07/2019] [Indexed: 10/25/2022] Open
Abstract
Purpose We present our results in the treatment of brain metastases (BM) from ovarian cancer using Gamma Knife Radiosurgery (GKRS) over the last 25 years in a single institution. Background Gamma Knife Radiosurgery has become increasingly important in the management of brain metastases from ovarian cancer due to improving results from systemic disease and the need for better outcomes. Material and methods The medical records of 9 patients with brain metastases from ovarian cancer treated with GKRS between 1993 and 2018 were reviewed. Median age at first treatment was 57 years (range 39-76). Forty-two brain metastases were treated with 16 procedures. Median tumor volume was 1.8cc ranging from 0.2 to 30.3cc (there were five patients with a tumor volume exceeding 10cc). Median prescription dose was 16 Gy. Results Using Kaplan Meier estimates, the median OS after diagnosis was 48.1 months and the median OS after GKRS was 10.6 months (ranging from 2.5 to 81 months). The Kaplan Meier survival rates were 31.3%, and 6.5% at 2 and 5 years after GKRS, respectively. Treatment procedure was well tolerated and no patient presented with acute or chronic toxicity. Two of 9 patients had a tumor requiring retreatment (local control of 95% 40/42). Two out of the 7 patients evaluated for cause of death expired due to progression of brain metastases and the remaining ones died of systemic disease with brain control. Conclusions GKRS for BM from ovarian cancer is a safe and effective modality. Our findings are in agreement with the recent literature indicating that women with brain metastases from ovarian cancer will benefit with radiosurgery and may achieve long term survival with brain control.
Collapse
Affiliation(s)
| | - Beatriz E Amendola
- Innovative Cancer Institute, 5995 SW 71 Street, South Miami, FL 33143, United States
| | - Paul F Martinez
- Innovative Cancer Institute, 5995 SW 71 Street, South Miami, FL 33143, United States
| | - Aizik Wolf
- Miami Neuroscience Center, 6129 SW 70 Street, South Miami, FL 33143, United States
| | - Sammie R Coy
- Miami Neuroscience Center, 6129 SW 70 Street, South Miami, FL 33143, United States
| | - Marco Amendola
- Innovative Cancer Institute, 5995 SW 71 Street, South Miami, FL 33143, United States
| |
Collapse
|
47
|
Macchia G, Lazzari R, Colombo N, Laliscia C, Capelli G, D'Agostino GR, Deodato F, Maranzano E, Ippolito E, Ronchi S, Paiar F, Scorsetti M, Cilla S, Ingargiola R, Huscher A, Cerrotta AM, Fodor A, Vicenzi L, Russo D, Borghesi S, Perrucci E, Pignata S, Aristei C, Morganti AG, Scambia G, Valentini V, Jereczek-Fossa BA, Ferrandina G. A Large, Multicenter, Retrospective Study on Efficacy and Safety of Stereotactic Body Radiotherapy (SBRT) in Oligometastatic Ovarian Cancer (MITO RT1 Study): A Collaboration of MITO, AIRO GYN, and MaNGO Groups. Oncologist 2019; 25:e311-e320. [PMID: 32043791 DOI: 10.1634/theoncologist.2019-0309] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/23/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Recent studies have reported improvement of outcomes (progression-free survival, overall survival, and prolongation of androgen deprivation treatment-free survival) with stereotactic body radiotherapy (SBRT) in non-small cell lung cancer and prostate cancer. The aim of this retrospective, multicenter study (MITO RT-01) was to define activity and safety of SBRT in a very large, real-world data set of patients with metastatic, persistent, and recurrent ovarian cancer (MPR-OC). MATERIALS AND METHODS The endpoints of the study were the rate of complete response (CR) to SBRT and the 24-month actuarial local control (LC) rate on "per-lesion" basis. The secondary endpoints were acute and late toxicities and the 24-month actuarial late toxicity-free survival. Objective response rate (ORR) included CR and partial response (PR). Clinical benefit (CB) included ORR and stable disease (SD). Toxicity was evaluated by the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC) and Common Terminology Criteria for Adverse Events (CTCAE) scales, according to center policy. Logistic and Cox regression were used for the uni- and multivariate analysis of factors predicting clinical CR and actuarial outcomes. RESULTS CR, PR, and SD were observed in 291 (65.2%), 106 (23.8%), and 33 (7.4%) lesions, giving a rate of CB of 96.4%. Patient aged ≤60 years, planning target volume (PTV) ≤18 cm3 , lymph node disease, and biologically effective dose α/β10 > 70 Gy were associated with higher chance of CR in the multivariate analysis. With a median follow-up of 22 months (range, 3-120), the 24-month actuarial LC rate was 81.9%. Achievement of CR and total dose >25 Gy were associated with better LC rate in the multivariate analysis. Mild toxicity was experienced in 54 (20.7%) patients; of 63 side effects, 48 were grade 1, and 15 were grade 2. The 24-month late toxicity-free survival rate was 95.1%. CONCLUSIONS This study confirms the activity and safety of SBRT in patients with MPR-OC and identifies clinical and treatment parameters able to predict CR and LC rate. IMPLICATIONS FOR PRACTICE This study aimed to define activity and safety of stereotactic body radiotherapy (SBRT) in a very large, real life data set of patients with metastatic, persistent, recurrent ovarian cancer (MPR-OC). Patient age <60 years, PTV <18 cm3 , lymph node disease, and biologically effective dose α/β10 >70 Gy were associated with higher chance of complete response (CR). Achievement of CR and total dose >25 Gy were associated with better local control (LC) rate. Mild toxicity was experienced in 20.7% of patients. In conclusion, this study confirms the activity and safety of SBRT in MPR-OC patients and identifies clinical and treatment parameters able to predict CR and LC rate.
Collapse
Affiliation(s)
- Gabriella Macchia
- Fondazione "Giovanni Paolo II," Unità Operativa di Radioterapia, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | - Roberta Lazzari
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Nicoletta Colombo
- Division of Medical Gynecologic Oncology, IEO European Institute of Oncology, IRCCS and University of Milan-Bicocca, Milan, Italy
| | - Concetta Laliscia
- Department of Translational Medicine, Division of Radiation Oncology, University of Pisa, Pisa, Italy
| | - Giovanni Capelli
- Department of Human Sciences, Society and Health, University of Cassino and Southern Lazio, Cassino, Italy
| | - Giuseppe Roberto D'Agostino
- Department of Radiotherapy and Radiosurgery, Humanitas Clinical and Research Hospital, IRCSS, Rozzano, Milan, Italy
| | - Francesco Deodato
- Fondazione "Giovanni Paolo II," Unità Operativa di Radioterapia, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | | | - Edy Ippolito
- Department of Radiation Oncology, Campus Bio-Medico University, Rome, Italy
| | - Sara Ronchi
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Fabiola Paiar
- Department of Translational Medicine, Division of Radiation Oncology, University of Pisa, Pisa, Italy
| | - Marta Scorsetti
- Department of Radiotherapy and Radiosurgery, Humanitas Clinical and Research Hospital, IRCSS, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Rozzano, Milano, Italy
| | - Savino Cilla
- Fondazione "Giovanni Paolo II," Unità Operativa di Fisica Medica, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | - Rossana Ingargiola
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Alessandra Huscher
- U.O. di Radioterapia Oncologica "Guido Berlucchi," Fondazione Poliambulanza, Brescia, Italy
| | - Anna Maria Cerrotta
- Radiotherapy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrei Fodor
- Department of Radiation Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Lisa Vicenzi
- Department of Oncology and Radiotherapy, Azienda Ospedaliera Universitaria Ospedali Riuniti, Ancona, Italy
| | | | - Simona Borghesi
- Radiotherapy Department, Azienda USL Toscana sud est, San Donato Hospital-Arezzo, Italy
| | - Elisabetta Perrucci
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Sandro Pignata
- Istituto Nazionale Tumori di Napoli, Fondazione Pascale IRCCS, Naples, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Alessio Giuseppe Morganti
- Department of Experimental, Diagnostic and Specialty Medicine - DIMES, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Giovanni Scambia
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Unità Operativa Complessa Ginecologia Oncologica, Dipartimento per la Salute della Donna e del Bambino e della Salute Pubblica, Roma, Italy
- Università Cattolica del Sacro Cuore, Istituto di Ginecologia e Ostetricia, Roma, Italy
| | - Vincenzo Valentini
- Fondazione "Giovanni Paolo II," Unità Operativa di Radioterapia, Università Cattolica del Sacro Cuore, Campobasso, Italy
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Unità Operativa Complessa di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Roma, Italy
- Università Cattolica del Sacro Cuore, Istituto di Radiologia, Roma, Italy
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Gabriella Ferrandina
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Unità Operativa Complessa Ginecologia Oncologica, Dipartimento per la Salute della Donna e del Bambino e della Salute Pubblica, Roma, Italy
- Università Cattolica del Sacro Cuore, Istituto di Ginecologia e Ostetricia, Roma, Italy
| |
Collapse
|
48
|
Park Y, Chui MH, Suryo Rahmanto Y, Yu ZC, Shamanna RA, Bellani MA, Gaillard S, Ayhan A, Viswanathan A, Seidman MM, Franco S, Leung AKL, Bohr VA, Shih IM, Wang TL. Loss of ARID1A in Tumor Cells Renders Selective Vulnerability to Combined Ionizing Radiation and PARP Inhibitor Therapy. Clin Cancer Res 2019; 25:5584-5594. [PMID: 31196855 DOI: 10.1158/1078-0432.ccr-18-4222] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/13/2019] [Accepted: 06/10/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Somatic inactivating mutations in ARID1A, a component of the SWI/SNF chromatin remodeling complex, are detected in various types of human malignancies. Loss of ARID1A compromises DNA damage repair. The induced DNA damage burden may increase reliance on PARP-dependent DNA repair of cancer cells to maintain genome integrity and render susceptibility to PARP inhibitor therapy.Experimental Design: Isogenic ARID1A-/- and wild-type cell lines were used for assessing DNA damage response, DNA compactness, and profiling global serine/threonine phosphoproteomic in vivo. A panel of inhibitors targeting DNA repair pathways was screened for a synergistic antitumor effect with irradiation in ARID1A-/- tumors. RESULTS ARID1A-deficient endometrial cells exhibit sustained levels in DNA damage response, a result further supported by in vivo phosphoproteomic analysis. Our results show that ARID1A is essential for establishing an open chromatin state upon DNA damage, a process required for recruitment of 53BP1 and RIF1, key mediators of non-homologous end-joining (NHEJ) machinery, to DNA lesions. The inability of ARID1A-/- cells to mount NHEJ repair results in a partial cytotoxic response to radiation. Small-molecule compound screens revealed that PARP inhibitors act synergistically with radiation to potentiate cytotoxicity in ARID1A-/- cells. Combination treatment with low-dose radiation and olaparib greatly improved antitumor efficacy, resulting in long-term remission in mice bearing ARID1A-deficient tumors. CONCLUSIONS ARID1A-deficient cells acquire high sensitivity to PARP inhibition after exposure to exogenously induced DNA breaks such as ionizing radiation. Our findings suggest a novel biologically informed strategy for treating ARID1A-deficient malignancies.
Collapse
Affiliation(s)
- Youngran Park
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - M Herman Chui
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yohan Suryo Rahmanto
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zheng-Cheng Yu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Raghavendra A Shamanna
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, Maryland
| | - Marina A Bellani
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, Maryland
| | - Stephanie Gaillard
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Gynecology/Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ayse Ayhan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology, Seirei Mikatahara General Hospital, Hamamatsu, Japan.,Hiroshima University School of Medicine, Hiroshima, Japan
| | - Akila Viswanathan
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael M Seidman
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, Maryland
| | - Sonia Franco
- Department of Gynecology/Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anthony K L Leung
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, Maryland
| | - Ie-Ming Shih
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,Richard W. TeLinde Gynecologic Pathology Research Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Richard W. TeLinde Gynecologic Pathology Research Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
49
|
Iorio GC, Martini S, Arcadipane F, Ricardi U, Franco P. The role of radiotherapy in epithelial ovarian cancer: a literature overview. Med Oncol 2019; 36:64. [PMID: 31165334 DOI: 10.1007/s12032-019-1287-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022]
Abstract
Ovarian cancer (OC) accounts for 3% of all cancer in women and for 5% of all cancer-related deaths. Epithelial Ovarian Cancer (EOC) is a radiosensitive malignancy with a poor prognosis. In the pre-chemotherapy era, radiation therapy (RT) delivered to the abdominopelvic region (whole abdominal irradiation, WAI) has historically played a role in the adjuvant and consolidation setting. Specific cluster of patients with early-stage disease and definite histologies may take advantage of RT. Platinum-based chemotherapy (CT) has replaced RT and plays a major role in most of the clinical settings. Radiation Therapy for palliation is recommended in patients with localized symptoms. Nevertheless, modern RT represents a reliable treatment option, with a mild toxicity profile, particularly effective for oligo-recurrent or progressive disease. The present literature review aims to highlight the historical role of RT in EOC, the actual lines of evidence, and the future perspectives.
Collapse
Affiliation(s)
- Giuseppe Carlo Iorio
- Department of Oncology, Radiation Oncology, School of Medicine, University of Turin, Via Genova 3, 10126, Turin, Italy
| | - Stefania Martini
- Department of Oncology, Radiation Oncology, School of Medicine, University of Turin, Via Genova 3, 10126, Turin, Italy
| | - Francesca Arcadipane
- Department of Oncology, Radiation Oncology, AOU Citta' della Salute e della Scienza, Turin, Italy
| | - Umberto Ricardi
- Department of Oncology, Radiation Oncology, School of Medicine, University of Turin, Via Genova 3, 10126, Turin, Italy
| | - Pierfrancesco Franco
- Department of Oncology, Radiation Oncology, School of Medicine, University of Turin, Via Genova 3, 10126, Turin, Italy.
| |
Collapse
|
50
|
Ghisoni E, Giannone G, Tuninetti V, Genta S, Scotto G, Aglietta M, Sangiolo D, Mittica G, Valabrega G. Veliparib: a new therapeutic option in ovarian cancer? Future Oncol 2019; 15:1975-1987. [DOI: 10.2217/fon-2018-0883] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The role of poly ADP ribose polymerase inhibitors in ovarian cancer is rapidly evolving. Three different poly ADP ribose polymerase inhibitors (olaparib, niraparib and rucaparib) have been already approved as maintenance after response to platinum-based chemotherapy; two of them (olaparib and rucaparib) also as single agents. Veliparib, a novel PARPI, showed promising results in preclinical and early clinical settings. The aim of this review is to discuss veliparib’s mechanisms of action, to provide a clinical update on its safety and activity in ovarian cancer, and to highlight future perspectives for its optimal use. Veliparib favorable toxicity profile encourages its use either as monotherapy or in combination. Its peculiar neuroprotective and radio-sensitizing effect warrant further investigation.
Collapse
Affiliation(s)
- Eleonora Ghisoni
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Gaia Giannone
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Valentina Tuninetti
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Sofia Genta
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Giulia Scotto
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Massimo Aglietta
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Dario Sangiolo
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Gloria Mittica
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
| | - Giorgio Valabrega
- Candiolo Cancer Institute FPO/IRCCS, Strada provinciale 142 km 3.95, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Torino, Italy
| |
Collapse
|