1
|
Zhang Y, Fang H, Wang X, Wang H, Pan G, Chen J. The Efficacy and Safety of Pazopanib Plus Chemotherapy in Treating Recurrent or Persistent Ovarian Cancer: A Systematic Review and Meta-Analysis. Am J Clin Oncol 2023; 46:254-262. [PMID: 36877187 PMCID: PMC10205118 DOI: 10.1097/coc.0000000000000999] [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: 03/07/2023]
Abstract
BACKGROUND Patients with recurrent or persistent ovarian cancer often have poor prognoses, and their optimal treatment regimen remains unclear. Inhibition of angiogenesis is a valuable strategy for treating ovarian cancer, and the drug pazopanib is a potent, multitarget tyrosine kinase inhibitor. However, treatment with pazopanib in combination with chemotherapy remains controversial. We performed a systematic review and meta-analysis to clarify the efficacy and side effects of pazopanib combined with chemotherapy in the treatment of advanced ovarian cancer. METHODS The PubMed, Embase, and Cochrane databases were systematically searched for relevant randomized controlled trials published up to September 2, 2022. The primary outcomes of eligible studies included overall response rate (ORR), disease control rate, 1-year progression-free survival (PFS) rate, 2-year PFS rate, 1-year overall survival (OS) rate, 2-year OS rate, and adverse events. RESULT Outcomes from a total of 518 recurrent or persistent ovarian cancer patients from 5 studies were analyzed in this systematic review. Pooled results showed that pazopanib plus chemotherapy, when compared with chemotherapy alone, significantly improved the ORR (pooled risk ratio=1.400; 95% CI, 1.062-1.846; P = 0.017) but not the disease control rate, 1-year PFS, 2-year PFS, 1-year OS, or 2-year OS. Moreover, pazopanib increased the risk of neutropenia, hypertension, fatigue, and liver dysfunction. CONCLUSION Pazopanib plus chemotherapy improved patient ORR but did not improve survival; it also increased the occurrence of several adverse events. Further large-sample clinical trials are needed to verify these results to guide pazopanib use in patients with ovarian cancer.
Collapse
|
2
|
Wang Y, Luo X, Wu N, Liao Q, Wang J. SRC-3/TRAF4 facilitates ovarian cancer development by activating the PI3K/AKT signaling pathway. Med Oncol 2023; 40:76. [PMID: 36625999 PMCID: PMC9831961 DOI: 10.1007/s12032-022-01944-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/26/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Ovarian cancer is the seventh most common cancer in women, and it causes many deaths in women worldwide. Patients with ovarian cancer have a poor prognosis and low survival rate. This study aimed to explore the role of the SRC-3/TRAF4/PI3K/AKT pathway in ovarian cancer development. METHODS SRC-3 and TRAF4 expression in ovarian cancer cell lines were assessed using qRT-PCR and western-blotting. The expression of SRC-3 and TRAF4 in ovarian cancer cells was downregulated by transient transfection with sh-RNAs. An MTT assay was performed to evaluate cell proliferation. Cell migration and invasion were measured using a Transwell assay. Cell stemness was detected using a cell spheroidization assay and western blotting. The expression levels of stem cell factors and PI3K/AKT pathway proteins were determined by qRT-PCR and western blot analysis. RESULTS SRC-3 and TRAF4 were upregulated in ovarian cancer cell lines. TRAF4 is a downstream factor of SRC-3, and the protein level of TRAF4 was regulated by SRC-3. SRC-3 knockdown reduced TRAF4 expression. Silencing SRC-3 or TRAF4 inhibited cell proliferation, migration, and invasion, as well as the expression of stem cell factors. Furthermore, sh-TRAF4 as well as treatment with LY294002, the PI3K/Akt inhibitor, inhibited the phosphorylation of Akt and PI3K, thus repressing the activation of PI3K/AKT signaling pathway in ovarian cancer cell lines. However, TRAF4 overexpression reversed the effect of SRC-3 silencing on cell proliferation, migration, invasion, and stemness. CONCLUSION Our study demonstrated that SRC-3/TRAF4 promotes ovarian cancer cell growth, migration, invasion, and stemness by activating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Ying Wang
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xia Luo
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Nayiyuan Wu
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jing Wang
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
3
|
Wang H, Wu M, Liu H, Zhou H, Zhao Y, Geng Y, Jiang B, Zhang K, Zhang B, Han Z, Du X. Comparison of the Efficacy and Safety of PARP Inhibitors as a Monotherapy for Platinum-Sensitive Recurrent Ovarian Cancer: A Network Meta-Analysis. Front Oncol 2021; 11:785102. [PMID: 34900739 PMCID: PMC8652073 DOI: 10.3389/fonc.2021.785102] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
Background The present COVID-19 pandemic has tended toward normality. To provide convenient, safe, and effective home treatment programs for patients with recurrent ovarian cancer (ROC), the clinical efficacy and safety of poly (ADP-ribose) polymerase inhibitor (PARPi) (including olaparib, niraparib, and rucaparib) monotherapy as a maintenance treatment for platinum-sensitive ROC were systematically evaluated. Methods Numerous electronic databases were systematically searched for randomized controlled trials (RCTs) of PARPi maintenance treatment for ROC that were published before June 2021. The primary endpoints were overall survival (OS) and progression-free survival (PFS), and the secondary endpoint was grade 3-4 adverse effects (AEs). After data extraction and the quality evaluation of the included studies, Bayesian network meta-analysis (NMA) was performed using R software. The ability of each treatment was ranked using the surface under the cumulative ranking (SUCRA) curve. Results The analysis included five studies and 1390 patients. The NMA results demonstrated that compared with the placebo, olaparib and niraparib exhibited significant benefits in the gBRCA-mutated population, and respectively reduced the risk of death by 31% (HR = 0.69, 95% CI: 0.53-0.90) and 34% (HR = 0.66, 95% CI: 0.44-0.99). Olaparib, niraparib, and rucaparib were all found to be very effective in prolonging PFS in patients with ROC. All three PARPi treatments increased the number of grade 3-4 AEs in patients with ROC as compared with the placebo. Conclusions Overall, olaparib and niraparib maintenance treatment can significantly prolong the OS of patients with gBRCA mutations. Furthermore, the three investigated PARPi monotherapy maintenance treatments can prolong PFS regardless of BRCA mutation status. Although the incidence of AEs in the treatment groups was found to be significantly higher than that in the placebo group, the patients in the treatment group tolerated the treatment. Home oral PARPi treatment can balance tumor treatment and pandemic prevention and control, and is the most convenient, safe, and effective home treatment method available against the background of the current COVID-19 pandemic. Systematic Review Registration https://inplasy.com/inplasy-2021-6-0033/.
Collapse
Affiliation(s)
- Hongmei Wang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Meng Wu
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Haonan Liu
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Hang Zhou
- Department of Hematology, The First People's Hospital of Lianyungang, Jiangsu, China
| | - Yang Zhao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Yifan Geng
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Bo Jiang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Kai Zhang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Bo Zhang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Zhengxiang Han
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Xiuping Du
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| |
Collapse
|
4
|
Keyvani V, Farshchian M, Esmaeili SA, Yari H, Moghbeli M, Nezhad SRK, Abbaszadegan MR. Ovarian cancer stem cells and targeted therapy. J Ovarian Res 2019; 12:120. [PMID: 31810474 PMCID: PMC6896744 DOI: 10.1186/s13048-019-0588-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Ovarian cancer has the highest ratio of mortality among gynecologic malignancies. Chemotherapy is one of the most common treatment options for ovarian cancer. However, tumor relapse in patients with advanced tumor stage is still a therapeutic challenge for its clinical management. MAIN BODY Therefore, it is required to clarify the molecular biology and mechanisms which are involved in chemo resistance to improve the survival rate of ovarian cancer patients. Cancer stem cells (CSCs) are a sub population of tumor cells which are related to drug resistance and tumor relapse. CONCLUSION In the present review, we summarized the recent findings about the role of CSCs in tumor relapse and drug resistance among ovarian cancer patients. Moreover, we focused on the targeted and combinational therapeutic methods against the ovarian CSCs.
Collapse
Affiliation(s)
- Vahideh Keyvani
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Moein Farshchian
- Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Bu‐Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Hadi Yari
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology, Tehran, Iran
| | - Meysam Moghbeli
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | | | | |
Collapse
|
5
|
Feng Y, Huang H, Wan T, Zhang C, Tong C, Liu J. Comparison of PARPis with Angiogenesis Inhibitors and Chemotherapy for Maintenance in Ovarian Cancer: A Network Meta-Analysis. Adv Ther 2019; 36:3368-3380. [PMID: 31599396 DOI: 10.1007/s12325-019-01106-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Seventy-five percent of ovarian cancer would relapse within 18-28 months after platinum-base chemotherapy. Evidence suggests that maintenance chemotherapy is effective in prolonging remission. Recent target therapies such as poly(ADP-ribose) polymerase inhibitors (PARPis) and angiogenesis inhibitors (AIs) are known to ease burden and recurrence of ovarian cancer. There is limited data for head-to-head comparison of PARPis, AIs, and chemotherapeutic agents (CTAs) as maintenance treatment. This network meta-analysis thus assessed the effectiveness and toxicity of these three maintenance therapies in patients with ovarian cancer. METHODS We searched relevant sources (PubMed, EMBASE) to identify randomized controlled trials assessing efficacy and safety of maintenance therapy in patients with ovarian cancer. Primary outcome was progression-free survival (PFS) as assessed by blinded review; safety and tolerability were secondary outcomes. A network meta-analysis to compare three drug classes was performed using statistical software R. RESULTS We included 24 trials (11,366 patients) assessing efficacy and safety of PARPis (n = 4), AIs (n = 12), and CTAs (n = 8). PARPis [hazard ratio (HR) 0.64; 95% credible intervals (CrI) 0.55-0.73] and AIs (HR 0.87; 95% CrI 0.81-0.93) showed significant improvement in PFS compared to placebo but not CTA (HR 1.00; 95% CrI 0.86-1.15). PARPis showed significant improvement in PFS compared to AIs (HR 0.73; 95% CrI 0.63-0.86) and CTA (HR 0.64; 95% CrI 0.52-0.78). Adverse events (AEs) leading to treatment discontinuation and dose reduction were lower in PARPis [incidence rate ratio (IRR) 0.60; CrI 0.31-1.18 and IRR 0.73, 95% CrI 0.50-1.06, respectively] compared to AIs, but the differences were not significant. CONCLUSION PARPi as maintenance treatment improved PFS in ovarian cancer and was relatively safer in terms of implications caused by AEs when compared to AIs. This network meta-analysis provides valuable evidence and significant insights into treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yanling Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - He Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ting Wan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Chuyao Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Chongjie Tong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jihong Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
6
|
Colon-Echevarria CB, Lamboy-Caraballo R, Aquino-Acevedo AN, Armaiz-Pena GN. Neuroendocrine Regulation of Tumor-Associated Immune Cells. Front Oncol 2019; 9:1077. [PMID: 31737559 PMCID: PMC6828842 DOI: 10.3389/fonc.2019.01077] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Mounting preclinical and clinical evidence continues to support a role for the neuroendocrine system in the modulation of tumor biology and progression. Several studies have shown data supporting a link between chronic stress and cancer progression. Dysregulation of the sympathetic nervous system (SNS) and the hypothalamic-pituitary-adrenal (HPA) axis has been implicated in promoting angiogenesis, tumor cell proliferation and survival, alteration of the immune response and exacerbating inflammatory networks in the tumor microenvironment. Here, we review how SNS and HPA dysregulation contributes to disturbances in immune cell populations, modifies cancer biology, and impacts immunotherapy response. We also highlight several interventions aimed at circumventing the adverse effects stress has on cancer patients.
Collapse
Affiliation(s)
- Claudia B Colon-Echevarria
- Division of Pharmacology, Department of Basic Sciences, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States
| | - Rocio Lamboy-Caraballo
- Division of Pharmacology, Department of Basic Sciences, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States
| | - Alexandra N Aquino-Acevedo
- Division of Pharmacology, Department of Basic Sciences, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States
| | - Guillermo N Armaiz-Pena
- Division of Pharmacology, Department of Basic Sciences, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States.,Divisions of Cancer Biology and Women's Health, Ponce Research Institute, Ponce, PR, United States
| |
Collapse
|
7
|
Wu L, Zhong L. Budget impact analysis of niraparib and olaparib for maintenance treatment of platinum-sensitive, recurrent ovarian cancer in the US. J Med Econ 2019; 22:187-195. [PMID: 30522378 DOI: 10.1080/13696998.2018.1557199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
AIMS This study aimed to evaluate the budget impact of niraparib and olaparib in patients with platinum-sensitive, recurrent ovarian cancer from a US third party payer perspective. MATERIALS AND METHODS A budget impact model was constructed to assess the additional per member per month (PMPM) costs associated with the introduction of niraparib and olaparib, two poly ADP-ribose polymerase ribose polymerase (PARP) inhibitors recently approved to be used in platinum-sensitive, recurrent ovarian cancer patients with and without a gBRCA mutation. The model assessed both pharmacy costs and medical costs. Pharmacy costs included adjusted drug costs, coinsurance, and dispensing fees. Medical costs included costs associated with disease monitoring and management of adverse events from the treatment. Epidemiological data from the literature were used to estimate the target population size. The analysis used 1-year time frame, and patients were assumed on treatment until disease progression or death. All costs were computed in 2017 USD. One-way sensitivity analyses were conducted to evaluate the model robustness. RESULTS In a hypothetical plan of 1,000,000 members, 206 patients were estimated to be potential candidates for niraparib or olaparib maintenance treatment after applying all epidemiological parameters. At listed 30-day supply WAC prices of $14,750 for niraparib and $13,482 for olaparib, budget impacts of these two drugs were $0.169 PMPM and $0.156 PMPM, respectively, most of which were contributed by pharmacy costs. Sensitivity analyses suggested that assumptions around market share, platinum-sensitive rate after first treatment, and WAC prices affected results the most. LIMITATIONS In this model, it was assumed that adopting niraparib and olaparib would not affect utilization of existing medications. Also, the estimated clinical parameters from clinical trials could differ from real-world data.
Collapse
Affiliation(s)
- Lei Wu
- a Irma Lerma Rangel College of Pharmacy , Texas A&M University , College Station , TX , USA
| | - Lixian Zhong
- a Irma Lerma Rangel College of Pharmacy , Texas A&M University , College Station , TX , USA
| |
Collapse
|
8
|
Villanucci A, Tavella K, Vannini L, Rossi V, Nobili S, Amunni G, Mazzei T, Mini E. Trabectedin in combination with pegylated liposomal doxorubicin in patients with ovarian tumors. Mol Clin Oncol 2018; 9:635-639. [DOI: 10.3892/mco.2018.1729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 09/06/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Alessandro Villanucci
- Department of Oncology, Hospital‑University Agency of Careggi, I‑50134 Florence, Italy
| | - Ketty Tavella
- Department of Oncology, Hospital‑University Agency of Careggi, I‑50134 Florence, Italy
| | - Laura Vannini
- Department of Oncology, University of Florence, I‑50134 Florence, Italy
| | - Virginia Rossi
- Department of Oncology, University of Florence, I‑50134 Florence, Italy
| | - Stefania Nobili
- Department of Oncology, University of Florence, I‑50134 Florence, Italy
| | - Gianni Amunni
- Department of Oncology, University of Florence, I‑50134 Florence, Italy
| | - Teresita Mazzei
- Department of Oncology, University of Florence, I‑50134 Florence, Italy
| | - Enrico Mini
- Department of Oncology, University of Florence, I‑50134 Florence, Italy
| |
Collapse
|
9
|
Cortez AJ, Tudrej P, Kujawa KA, Lisowska KM. Advances in ovarian cancer therapy. Cancer Chemother Pharmacol 2018; 81:17-38. [PMID: 29249039 PMCID: PMC5754410 DOI: 10.1007/s00280-017-3501-8] [Citation(s) in RCA: 398] [Impact Index Per Article: 56.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/11/2017] [Indexed: 02/06/2023]
Abstract
Epithelial ovarian cancer is typically diagnosed at an advanced stage. Current state-of-the-art surgery and chemotherapy result in the high incidence of complete remissions; however, the recurrence rate is also high. For most patients, the disease eventually becomes a continuum of symptom-free periods and recurrence episodes. Different targeted treatment approaches and biological drugs, currently under development, bring the promise of turning ovarian cancer into a manageable chronic disease. In this review, we discuss the current standard in the therapy for ovarian cancer, major recent studies on the new variants of conventional therapies, and new therapeutic approaches, recently approved and/or in clinical trials. The latter include anti-angiogenic therapies, polyADP-ribose polymerase (PARP) inhibitors, inhibitors of growth factor signaling, or folate receptor inhibitors, as well as several immunotherapeutic approaches. We also discuss cost-effectiveness of some novel therapies and the issue of better selection of patients for personalized treatment.
Collapse
Affiliation(s)
- Alexander J Cortez
- Maria Skłodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-100, Poland
| | - Patrycja Tudrej
- Maria Skłodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-100, Poland
| | - Katarzyna A Kujawa
- Maria Skłodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-100, Poland
| | - Katarzyna M Lisowska
- Maria Skłodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-100, Poland.
| |
Collapse
|
10
|
Tremble LF, Forde PF, Soden DM. Clinical evaluation of macrophages in cancer: role in treatment, modulation and challenges. Cancer Immunol Immunother 2017; 66:1509-1527. [PMID: 28948324 PMCID: PMC11028704 DOI: 10.1007/s00262-017-2065-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/13/2017] [Indexed: 12/22/2022]
Abstract
The focus of immunotherapeutics has been placed firmly on anti-tumour T cell responses. Significant progress has been made in the treatment of both local and systemic malignancies, but low response rates and rising toxicities are limiting this approach. Advancements in the understanding of tumour immunology are opening up a new range of therapeutic targets, including immunosuppressive factors in the tumour microenvironment. Macrophages are a heterogeneous group of cells that have roles in innate and adaptive immunity and tissue repair, but become co-opted by tumours to support tumour growth, survival, metastasis and immunosuppression. Macrophages also support tumour resistance to conventional therapy. In preclinical models, interference with macrophage migration, macrophage depletion and macrophage re-education have all been shown to reduce tumour growth and support anti-tumour immune responses. Here we discuss the role of macrophages in prognosis and sensitivity to therapy, while examining the significant progress which has been made in modulating the behaviour of these cells in cancer patients.
Collapse
Affiliation(s)
- Liam Friel Tremble
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland.
| | - Patrick F Forde
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland
| | - Declan M Soden
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland
| |
Collapse
|
11
|
Borgoni S, Iannello A, Cutrupi S, Allavena P, D'Incalci M, Novelli F, Cappello P. Depletion of tumor-associated macrophages switches the epigenetic profile of pancreatic cancer infiltrating T cells and restores their anti-tumor phenotype. Oncoimmunology 2017; 7:e1393596. [PMID: 29308326 DOI: 10.1080/2162402x.2017.1393596] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/30/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDA) is characterized by a complex tumor microenvironment that supports its progression, aggressiveness and resistance to therapies. The delicate interplay between cancer and immune cells creates the conditions for PDA development, particularly due to the functional suppression of T cell anti-tumor effector activity. However, some of the mechanisms involved in this process are still poorly understood. In this study, we analyze whether the functional and epigenetic profile of T cells that infiltrate PDA is modulated by the microenvironment, and in particular by tumor-associated macrophages (TAMs). CD4 and CD8 T cells obtained from mice orthotopically injected with syngeneic PDA cells, and untreated or treated with Trabectedin, a cytotoxic drug that specifically targets TAMs, were sorted and analyzed by flow cytometry and characterized for their epigenetic profile. Assessment of cytokine production and the epigenetic profile of genes coding for IL10, T-bet and PD1 revealed that T cells that infiltrated PDA displayed activated Il10 promoter and repressed T-bet activity, in agreement with their regulatory phenotype (IL10high/IFNγlow, PD1high). By contrast, in Trabectedin-treated mice, PDA-infiltrating T cells displayed repressed Il10 and Pdcd1 and activated T-bet promoter activity, in accordance with their anti-tumor effector phenotype (IL10low/IFNγhigh), indicating a key role of TAMs in orchestrating functions of PDA-infiltrating T cells by modulating their epigenetic profile towards a pro-tumoral phenotype. These results suggest the targeting of TAMs as an efficient strategy to obtain an appropriate T cell anti-tumor immune response and open new potential combinations for PDA treatment.
Collapse
Affiliation(s)
- Simone Borgoni
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, via Nizza 52, Torino, Italy.,Center for Experimental Research and Medical Studies, University Hospital Città della Salute e della Scienza di Torino, via Santena 5, Torino, Italy
| | - Andrea Iannello
- Center for Molecular Systems Biology, University of Turin, Orbassano, Turin, Italy.,Dept. of Clinical and Biological Sciences, University of Turin, Orbassano, Turin, Italy
| | - Santina Cutrupi
- Center for Molecular Systems Biology, University of Turin, Orbassano, Turin, Italy.,Dept. of Clinical and Biological Sciences, University of Turin, Orbassano, Turin, Italy
| | - Paola Allavena
- Dept. Immunology and Inflammation, IRCCS-Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano (Milano), Italy
| | - Maurizio D'Incalci
- Dept. of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, Milan, Italy
| | - Francesco Novelli
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, via Nizza 52, Torino, Italy.,Center for Experimental Research and Medical Studies, University Hospital Città della Salute e della Scienza di Torino, via Santena 5, Torino, Italy.,Transplant Immunology Service, University Hospital Città della Salute e della Scienza di Torino, Turin, Italy.,Molecular Biotechnology Center, via Nizza 52, Torino, Italy
| | - Paola Cappello
- Dept. of Molecular Biotechnology and Health Sciences, University of Turin, via Nizza 52, Torino, Italy.,Center for Experimental Research and Medical Studies, University Hospital Città della Salute e della Scienza di Torino, via Santena 5, Torino, Italy.,Molecular Biotechnology Center, via Nizza 52, Torino, Italy
| |
Collapse
|
12
|
Rubio Pérez MJ. Effect of the Combination of Trabectedin and Pegylated Liposomal Doxorubicin in a BRCA2 Mutation Carrier with Recurrent Platinum-Sensitive Ovarian Cancer. Case Rep Oncol 2017. [PMID: 28626402 PMCID: PMC5471796 DOI: 10.1159/000475707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Introduction The current standard of care for ovarian cancer is optimal cytoreduction with adjuvant chemotherapy based on a platinum/taxane combination. Although the response rate to this therapy is high, most patients ultimately relapse. Response to second-line therapy and prognosis are linked to the platinum-free interval (PFI); when both improve, the PFI increases. As a result, there is an increasing interest in the PFI extension strategies including platinum-free combinations. Case Presentation A 50-year-old postmenopausal woman presented with ovarian serous carcinoma with peritoneal carcinomatosis. First-line neoadjuvant chemotherapy with carboplatin plus paclitaxel was initiated, followed by surgery and carboplatin plus paclitaxel chemotherapy. Eight months after the last cycle, CT revealed extensive supra- and infradiaphragmatic node involvement, and second-line chemotherapy was initiated with trabectedin and pegylated liposomal doxorubicin (PLD). Partial response was achieved and successfully maintained for 18 cycles. After the 18th cycle and a 25-month PFI, CT imaging evidenced disease progression. As the patient was a BRCA2 mutation carrier, third-line chemotherapy was initiated with carboplatin and gemcitabine every 3 weeks. After the third cycle, imaging confirmed complete response, which was maintained after the sixth and final cycle. Maintenance treatment with olaparib was initiated. At present – 6 months after the start of maintenance chemotherapy with olaparib – the patient is disease free. Conclusions Second-line chemotherapy with a nonplatinum combination – trabectedin plus PLD – was effective in a BRCA2 mutation carrier with recurrent partially platinum-sensitive ovarian cancer.
Collapse
|
13
|
Lindequist U. Marine-Derived Pharmaceuticals - Challenges and Opportunities. Biomol Ther (Seoul) 2016; 24:561-571. [PMID: 27795450 PMCID: PMC5098534 DOI: 10.4062/biomolther.2016.181] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/04/2016] [Accepted: 10/05/2016] [Indexed: 01/23/2023] Open
Abstract
Marine biosphere is the largest one of the earth and harbors an enormous number of different organisms. Living conditions differ fundamentally from those in terrestrial environment. The production of specific secondary metabolites is an important adaption mechanism of marine organisms to survive in the sea. These metabolites possess biological activities which make them interesting as possible drugs for human. The review presents sources, chemistry, production and pharmacology of FDA approved marine derived pharmaceuticals arranged according to their therapeutic indication. Four of the presently seven approved drugs are used for the treatment of cancer. Each another one is applicated for treatment of viral diseases, chronic pain and to lower triglyceride level in blood. Some other products are of interest in diagnostic and as experimental tools. Besides, this article describes challenges in drug development from marine sources, especially the supply problem.
Collapse
Affiliation(s)
- Ulrike Lindequist
- Department of Pharmaceutical Biology, Institute of Pharmacy, Ernst-Moritz-Arndt University of Greifswald, Greifswald D17489, Germany
| |
Collapse
|
14
|
Abstract
In this issue of the Chinese Journal of Cancer, European, American, and Chinese experts review the current management and future perspectives of epithelial ovarian cancer (EOC), the leading cause of gynecological cancer deaths. Although major advances have been made in understanding the cellular and molecular biology of this highly heterogeneous malignancy, the survival rate of women with EOC has changed little since the introduction of platinum-based treatment as a front-line therapy. The papers describe the progress in deciphering the molecular complexity of this disease and the newly available molecular-driven therapies, which have been applied by shifting trial designs toward restricting eligibility to specific subgroups of patients rather than testing agents in unselected populations. These new trial designs provide potential opportunities for improved efficacy in targeted populations. Given the molecular complexity of this disease, patient survival may be increased by searching for new molecular prognostic/predictive signatures as well as by translating the recent insight of microRNA involvement in EOC progression into new, targeted therapies. Particular attention has been given to the issue of fertility sparing for women affected by curable diseases.
Collapse
Affiliation(s)
- Delia Mezzanzanica
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano 20133, Italy.
| |
Collapse
|
15
|
Xyloketal B suppresses glioblastoma cell proliferation and migration in vitro through inhibiting TRPM7-regulated PI3K/Akt and MEK/ERK signaling pathways. Mar Drugs 2015; 13:2505-25. [PMID: 25913706 PMCID: PMC4413223 DOI: 10.3390/md13042505] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/03/2015] [Accepted: 04/08/2015] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma, the most common and aggressive type of brain tumors, has devastatingly proliferative and invasive characteristics. The need for finding a novel and specific drug target is urgent as the current approaches have limited therapeutic effects in treating glioblastoma. Xyloketal B is a marine compound obtained from mangrove fungus Xylaria sp. (No. 2508) from the South China Sea, and has displayed antioxidant activity and protective effects on endothelial and neuronal oxidative injuries. In this study, we used a glioblastoma U251 cell line to (1) explore the effects of xyloketal B on cell viability, proliferation, and migration; and (2) investigate the underlying molecular mechanisms and signaling pathways. MTT assay, colony formation, wound healing, western blot, and patch clamp techniques were employed. We found that xyloketal B reduced cell viability, proliferation, and migration of U251 cells. In addition, xyloketal B decreased p-Akt and p-ERK1/2 protein expressions. Furthermore, xyloketal B blocked TRPM7 currents in HEK-293 cells overexpressing TRPM7. These effects were confirmed by using a TRPM7 inhibitor, carvacrol, in a parallel experiment. Our findings indicate that TRPM7-regulated PI3K/Akt and MEK/ERK signaling is involved in anti-proliferation and migration effects of xyloketal B on U251 cells, providing in vitro evidence for the marine compound xyloketal B to be a potential drug for treating glioblastoma.
Collapse
|