1
|
Cetinkaya A, Kaya SI, Budak F, Ozkan SA. Current Analytical Methods for the Sensitive Assay of New-Generation Ovarian Cancer Drugs in Pharmaceutical and Biological Samples. Crit Rev Anal Chem 2024:1-17. [PMID: 38630637 DOI: 10.1080/10408347.2024.2339962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Ovarian cancer, which affects the female reproductive organs, is one of the most common types of cancer. Since this type of cancer has a high mortality rate from gynaecological cancers, the scientific community shows great interest in studies on its treatment. Chemotherapy, radiotherapy, and surgical treatment methods are used in its treatment. In the absence of targeted treatments in these treatment methods, side effects occur in patients, and patients show resistance to the drug. In addition, the underlying causes of ovarian cancer are still not fully known. The scientific world thinks that genetic factors, environmental conditions, and consumed foods may cause this cancer. The most important factor in the treatment of ovarian cancer is early diagnosis. Therefore, the drugs used in the treatment of ovarian cancer are platinum-based anticancer drugs. In addition to these drugs, the most preferred treatment method recently is targeted treatment approaches using poly(adenosine diphosphate ribose) polymerase (PARP) inhibitors. In this review, studies on the sensitive analysis of the treatment methods of these new-generation drugs used in the treatment of ovarian cancer have been comprehensively examined. In addition, the basic features, structural aspects, and biological data of analytical methods used in treatments with new-generation drugs are explained. Analytical studies carried out in the literature in recent years aim to show future developments in how these new-generation drugs are used today and to guide future studies by comprehensively examining and explaining the structure-activity relationship, mechanism of action, toxicity, and pharmacokinetic studies. Finally, in this study, the methods used in the analysis of drugs used in the treatment of ovarian cancer and the studies conducted between 2015 and 2023 were discussed in detail.
Collapse
Affiliation(s)
- Ahmet Cetinkaya
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
| | - S Irem Kaya
- Gulhane Faculty of Pharmacy, Department of Analytical Chemistry, University of Health Sciences, Ankara, Turkey
| | - Fatma Budak
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
- Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - Sibel A Ozkan
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
| |
Collapse
|
2
|
Targeting receptor tyrosine kinases in ovarian cancer: Genomic dysregulation, clinical evaluation of inhibitors, and potential for combinatorial therapies. Mol Ther Oncolytics 2023; 28:293-306. [PMID: 36911068 PMCID: PMC9999170 DOI: 10.1016/j.omto.2023.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Epithelial ovarian cancer (EOC) remains one of the leading causes of cancer-related deaths among women worldwide. Receptor tyrosine kinases (RTKs) have long been sought as therapeutic targets for EOC, as they are frequently hyperactivated in primary tumors and drive disease relapse, progression, and metastasis. More recently, these oncogenic drivers have been implicated in EOC response to poly(ADP-ribose) polymerase (PARP) inhibitors and epigenome-interfering agents. This evidence revives RTKs as promising targets for therapeutic intervention of EOC. This review summarizes recent studies on the role of RTKs in EOC malignancy and the use of their inhibitors for clinical treatment. Our focus is on the ERBB family, c-Met, and VEGFR, as they are linked to drug resistance and targetable using commercially available drugs. The importance of these RTKs and their inhibitors is highlighted by their impact on signal transduction and intratumoral heterogeneity in EOC and successful use as maintenance therapy in the clinic through suppression of the VEGF/VEGFR axis. Finally, the therapeutic potential of RTK inhibitors is discussed in the context of combinatorial targeting via co-inhibiting proliferative and anti-apoptotic pathways, epigenomic/transcriptional programs, and harnessing the efficacy of PARP inhibitors and programmed cell death 1/ligand 1 immune checkpoint therapies.
Collapse
|
3
|
Hartnett EG, Knight J, Radolec M, Buckanovich RJ, Edwards RP, Vlad AM. Immunotherapy Advances for Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12123733. [PMID: 33322601 PMCID: PMC7764119 DOI: 10.3390/cancers12123733] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022] Open
Abstract
Simple Summary The overall five-year survival rate in epithelial ovarian cancer is 44% and has only marginally improved in the past two decades. Despite an initial response to standard treatment consisting of chemotherapy and surgical removal of tumor, the lesions invariably recur, and patients ultimately die of chemotherapy resistant disease. New treatment modalities are needed in order to improve the prognosis of women diagnosed with ovarian cancer. One such modality is immunotherapy, which aims to boost the capacity of the patient’s immune system to recognize and attack the tumor cells. We performed a retrospective study to identify some of the most promising immune therapies for epithelial ovarian cancer. Special emphasis was given to immuno-oncology clinical trials. Abstract New treatment modalities are needed in order to improve the prognosis of women diagnosed with epithelial ovarian cancer (EOC), the most aggressive gynecologic cancer type. Most ovarian tumors are infiltrated by immune effector cells, providing the rationale for targeted approaches that boost the existing or trigger new anti-tumor immune mechanisms. The field of immuno-oncology has experienced remarkable progress in recent years, although the results seen with single agent immunotherapies in several categories of solid tumors have yet to extend to ovarian cancer. The challenge remains to determine what treatment combinations are most suitable for this disease and which patients are likely to benefit and to identify how immunotherapy should be incorporated into EOC standard of care. We review here some of the most promising immune therapies for EOC and focus on those currently tested in clinical trials.
Collapse
Affiliation(s)
- Erin G. Hartnett
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Julia Knight
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Mackenzy Radolec
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Ronald J. Buckanovich
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Robert P. Edwards
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Anda M. Vlad
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
- Correspondence:
| |
Collapse
|
4
|
Lindenblatt D, Terraneo N, Pellegrini G, Cohrs S, Spycher PR, Vukovic D, Béhé M, Schibli R, Grünberg J. Combination of lutetium-177 labelled anti-L1CAM antibody chCE7 with the clinically relevant protein kinase inhibitor MK1775: a novel combination against human ovarian carcinoma. BMC Cancer 2018; 18:922. [PMID: 30253737 PMCID: PMC6156869 DOI: 10.1186/s12885-018-4836-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 09/19/2018] [Indexed: 12/21/2022] Open
Abstract
Background Protein kinase inhibitors (PKIs) are currently tested in clinical studies (phase I-III) as an alternative strategy against (recurrent) ovarian cancer. Besides their anti-tumour efficacy, several PKIs have also shown radiosensitizing effects when combined with external beam radiation. Based on these results we asked if the addition of PKIs offers a therapeutic opportunity to improve radioimmunotherapy (RIT) against ovarian cancer. Five PKIs (alisertib, MK1775, MK2206, saracatinib, temsirolimus) were chosen for cytotoxicity screenings based on their current clinical trials in the treatment of ovarian cancer and their influence on cell cycle regulation and DNA damage repair pathways. We combined selected PKIs with 177Lu-labelled anti-L1CAM monoclonal antibody chCE7 for our investigations. Methods PKIs cytotoxicity was determined via cell colony-forming assays. Biomarker of DNA double-strand breaks (DSBs, γH2A.X) was analysed by western blot and fluorescence microscopy. Flow cytometric measurements were performed to evaluate levels of apoptosis based on mono- or combination treatments. The best combination was used for in vivo combination therapy studies in nude mice with SKOV3ip and IGROV1 human ovarian cancer xenografts. Bonferroni correction was used to determine statistical significance for multiple comparisons. Results The highest cytotoxicity against both cell lines was observed for MK1775 and alisertib. Combinations including 177Lu-labelled mAb chCE7 and MK1775 decreased 177Lu-DOTA-chCE7 IC60-values 14-fold, compared to 6-fold, when the radioimmunoconjugate was combined with alisertib. The most effective PKI MK1775 was further evaluated and demonstrated synergistic effects in combination with 177Lu-DOTA-chCE7 against IGROV1 cells. Significantly higher amounts of DSBs were detected in IGROV1 cells after combination (91%) compared to either treatment alone (MK1775: 52%; radioimmunoconjugate: 72%; p < 0.0125). Early-apoptosis was significantly enhanced in IGROV1 cells correlating with induced DSBs (177Lu-DOTA-chCE7: 8%, MK1775: 28%, 177Lu-DOTA-chCE7 + MK1775: 40%, p < 0.0125). Immunohistochemistry analysis of γH2A.X expression levels after therapy in SKOV3ip xenografts revealed a high sensitivity of the tumour cells to MK1775 and a high radioresistance. A prominent effect of tumour growth inhibition of the RIT and of the combination therapy was observed in vivo in a late stage IGROV1 xenograft model. Conclusions Our results warrant further evaluation of combination of MK1775 and radioimmunotherapy. Electronic supplementary material The online version of this article (10.1186/s12885-018-4836-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dennis Lindenblatt
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland
| | - Nastassja Terraneo
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland
| | - Giovanni Pellegrini
- Institut for Veterinary Pathology, University of Zurich, Zurich, Switzerland
| | - Susan Cohrs
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland
| | - Philipp René Spycher
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland
| | - David Vukovic
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland
| | - Martin Béhé
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zürich, Zurich, Switzerland
| | - Jürgen Grünberg
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland.
| |
Collapse
|
5
|
Targeting HER2 in ovarian and uterine cancers: Challenges and future directions. Gynecol Oncol 2014; 135:364-70. [DOI: 10.1016/j.ygyno.2014.09.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/28/2014] [Accepted: 09/03/2014] [Indexed: 01/22/2023]
|
6
|
Missaoui N, Abdelkarim SB, Ayachi M, Hmissa S, Yaacoubi MT. HER2 expression in ovarian mucinous carcinomas in Tunisia. Asian Pac J Cancer Prev 2014; 15:8121-5. [PMID: 25338994 DOI: 10.7314/apjcp.2014.15.19.8121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian mucinous carcinoma has a poor prognosis in advanced stages and a poor response to conventional chemotherapy. An efficient treatment is not yet available. We heere investigated HER2 expression and the potential for trastuzumab therapy in ovarian mucinous tumors. MATERIALS AND METHODS Immunohistochemistry was performed in formalin-fixed, paraffin-embedded tissue from 27 ovarian mucinous tumors including 14 carcinomas and 13 borderline tumors diagnosed in the Pathology Department, Farhet Hached Hospital, Sousse, between 1993 and 2013. The HercepTest (DAKO) was used for immunohistochemistry. RESULTS HER2 expression was observed in only one borderline tumor (7.7%) and in 14.3% of mucinous carcinomas of the ovary. CONCLUSIONS Our results suggest that trastuzumab therapy would be an option for patients with mucinous carcinoma when the tumor has HER2 overexpression.
Collapse
Affiliation(s)
- Nabiha Missaoui
- Research Unit03/UR/08-13, Cancer Epidemiology and Cytopathology in Tunisian Center, Faculty of Medicine, Sousse, Tunisia E-mail :
| | | | | | | | | |
Collapse
|
7
|
Lindenblatt D, Fischer E, Cohrs S, Schibli R, Grünberg J. Paclitaxel improved anti-L1CAM lutetium-177 radioimmunotherapy in an ovarian cancer xenograft model. EJNMMI Res 2014; 4:54. [PMID: 26116117 PMCID: PMC4452682 DOI: 10.1186/s13550-014-0054-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/26/2014] [Indexed: 12/18/2022] Open
Abstract
Background Today’s standard treatment of advanced-stage ovarian cancer, including surgery followed by a paclitaxel-platinum-based chemotherapy, is limited in efficacy. Recently, we could show that radioimmunotherapy (RIT) with 177Lu-labelled anti-L1 cell adhesion molecule (L1CAM) monoclonal antibody chCE7 is effective in ovarian cancer therapy. We investigated if the efficacy of anti-L1CAM RIT can be further improved by its combination with paclitaxel (PTX). Methods In vitro cell viability and cell cycle arrest of human ovarian cancer cells were assessed upon different treatment conditions. For therapy studies, nude mice (n = 8) were injected subcutaneously with IGROV1 human ovarian carcinoma cells and received a single dose of 6 MBq 177Lu-DOTA-chCE7 alone or in combination with 600 μg PTX (31.6 mg/kg). Tumour growth delay and survival were determined. To investigate whether PTX can influence the tumour uptake of the radioimmunoconjugates (RICs), a biodistribution study (n = 4) and SPECT/CT images were acquired 120 h post injections of 2 MBq 177Lu-DOTA-chCE7 alone or in combination with 600 μg PTX. Results Lu-DOTA-chCE7 in combination with PTX revealed a significantly decreased cell viability of ovarian carcinoma cells in vitro and was effective in a synergistic manner (combination index < 1). PTX increased the RIT efficacy by arresting cells in the radiosensitive G2/M phase of the cell cycle 24 h post treatment start. In vivo combination therapy including 177Lu-DOTA-chCE7 and PTX resulted in a significantly prolonged overall survival (55 days vs. 18 days/PTX and 29 days/RIT), without weight loss and/or signs of toxicity. Biodistribution studies revealed no significant difference in tumour uptakes of 177Lu-DOTA-chCE7 72 h post injection regardless of an additional PTX administration. Conclusions Combination of anti-L1CAM 177Lu-RIT with PTX is a more effective therapy resulting in a prolonged overall survival of human ovarian carcinoma-bearing nude mice compared with either monotherapy. The combination is promising for future clinical applications. Electronic supplementary material The online version of this article (doi:10.1186/s13550-014-0054-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dennis Lindenblatt
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland,
| | | | | | | | | |
Collapse
|
8
|
Lavoué V, Foucher F, Henno S, Bauville E, Catros V, Cabillic F, Levêque J. [Immunotherapy in epithelial ovarian carcinoma: hope and reality]. ACTA ACUST UNITED AC 2013; 43:198-210. [PMID: 24230482 DOI: 10.1016/j.jgyn.2013.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/14/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Epithelial ovarian carcinoma (EOC) has a worst prognosis with little progress in terms of survival for the last two decades. Immunology received little interest in EOC in the past, but now appears very important in the natural history of this cancer. This review is an EOC immunology state of art and focuses on the place of immunotherapy in future. MATERIAL AND METHODS A systematic review of published studies was performed. Medline baseline interrogation was performed with the following keywords: "Ovarian carinoma, immunotherapy, T-lymphocyte, regulator T-lymphocyte, dendritic cells, macrophage, antigen, chemotherapy, surgery, clinical trials". Identified publications (English or French) were assessed for the understanding of EOC immunology and the place of conventional treatment and immunotherapy strategy. RESULTS Intratumoral infiltration by immune cells is a strong prognotic factor in EOC. Surgery and chemotherapy in EOC decrease imunosuppression in patients. The antitumoral immunity is a part of the therapeutic action of surgery and chemotherapy. Until now, immunotherapy gave some disappointing results, but the new drugs that target the tolerogenic tumoral microenvironnement rise and give a new hope in the treatment of cancer. CONCLUSION Immunology controls the EOC natural history. The modulation of immunosuppressive microenvironment associated with the stimulation of antitumoral immunity could be the next revolution in the treatment of cancer.
Collapse
Affiliation(s)
- V Lavoué
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France; Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France.
| | - F Foucher
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - S Henno
- Service d'anatomo-pathologie, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - E Bauville
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - V Catros
- Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France; Service de biologie cellulaire, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - F Cabillic
- Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France; Service de biologie cellulaire, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - J Levêque
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France; Faculté de médecine, université de Rennes 1, 35000 Rennes, France
| |
Collapse
|
9
|
Song JX, Li FQ, Cao WL, Jia X, Shi LN, Lu JF, Ma CF, Kong QQ. Anti-Sp17 monoclonal antibody–doxorubicin conjugates as molecularly targeted chemotherapy for ovarian carcinoma. Target Oncol 2013; 9:263-72. [DOI: 10.1007/s11523-013-0293-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 07/15/2013] [Indexed: 10/26/2022]
|
10
|
Lavoué V, Thédrez A, Levêque J, Foucher F, Henno S, Jauffret V, Belaud-Rotureau MA, Catros V, Cabillic F. Immunity of human epithelial ovarian carcinoma: the paradigm of immune suppression in cancer. J Transl Med 2013; 11:147. [PMID: 23763830 PMCID: PMC3683338 DOI: 10.1186/1479-5876-11-147] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 05/27/2013] [Indexed: 12/21/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a significant cause of cancer-related mortality in women, and there has been no substantial decrease in the death rates due to EOC in the last three decades. Thus, basic knowledge regarding ovarian tumor cell biology is urgently needed to allow the development of innovative treatments for EOC. Traditionally, EOC has not been considered an immunogenic tumor, but there is evidence of an immune response to EOC in patients. Clinical data demonstrate that an antitumor immune response and immune evasion mechanisms are correlated with a better and lower survival, respectively, providing evidence for the immunoediting hypothesis in EOC. This review focuses on the immune response and immune suppression in EOC. The immunological roles of chemotherapy and surgery in EOC are also described. Finally, we detail pilot data supporting the efficiency of immunotherapy in the treatment of EOC and the emerging concept that immunomodulation aimed at counteracting the immunosuppressive microenvironment must be associated with immunotherapy strategies.
Collapse
Affiliation(s)
- Vincent Lavoué
- Lady Davis Institut, Jewish General Hospital, McGill University, Montreal QC H3T 1E2, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
The role of tumour-associated MUC1 in epithelial ovarian cancer metastasis and progression. Cancer Metastasis Rev 2013; 32:535-51. [DOI: 10.1007/s10555-013-9423-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
12
|
Sparholt MH, Høgdall CK, Nedergaard L, Heeran MCJ, Christensen IJ, Høgdall EVS. Is tissue CA125 expression in epithelial ovarian adenocarcinoma heterogenic? APMIS 2012; 121:38-44. [DOI: 10.1111/j.1600-0463.2012.02943.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 05/25/2012] [Indexed: 11/30/2022]
Affiliation(s)
- Morten H. Sparholt
- The Gynecologic Clinic; The Juliane Marie Centre; Rigshospitalet; University of Copenhagen; Copenhagen; Denmark
| | - Claus K. Høgdall
- The Gynecologic Clinic; The Juliane Marie Centre; Rigshospitalet; University of Copenhagen; Copenhagen; Denmark
| | - Lotte Nedergaard
- Department of Pathology; Rigshospitalet; University of Copenhagen; Copenhagen; Denmark
| | - Mel C. J. Heeran
- Department of Pathology; Molecular Unit; Herlev Hospital; University of Copenhagen; Copenhagen; Denmark
| | | | - Estrid V. S. Høgdall
- Department of Pathology; Molecular Unit; Herlev Hospital; University of Copenhagen; Copenhagen; Denmark
| |
Collapse
|
13
|
Abstract
Epithelial ovarian cancer (EOC) remains the most lethal gynecological malignancy despite several decades of progress in diagnosis and treatment. Taking advantage of the robust development of discovery and utility of prognostic biomarkers, clinicians and researchers are developing personalized and targeted treatment strategies. This review encompasses recently discovered biomarkers of ovarian cancer, the utility of published prognostic biomarkers for EOC (especially biomarkers related to angiogenesis and key signaling pathways), and their integration into clinical practice.
Collapse
Affiliation(s)
- Jie Huang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | |
Collapse
|
14
|
Hillig T, Thode J, Breinholt MF, Franzmann MB, Pedersen C, Lund F, Mygind H, Sölétormos G, Rudnicki M. Assessing HER2 amplification by IHC, FISH, and real-time polymerase chain reaction analysis (real-time PCR) following LCM in formalin-fixed paraffin embedded tissue from 40 women with ovarian cancer. APMIS 2012; 120:1000-7. [PMID: 23030524 PMCID: PMC3533780 DOI: 10.1111/j.1600-0463.2012.02929.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 05/07/2012] [Indexed: 12/21/2022]
Abstract
We compare HER2 receptor amplification analysis by immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and real-time polymerase chain reaction (real-time PCR) DNA copy-number assay following laser capture microdissection (LCM) in formalin-fixed paraffin embedded tissue from 40 women with verified ovarian cancer. We speculate that LCM should result in a more accurate assessment of HER2 amplification in our real-time PCR assay compared with IHC and FISH. HER2 overexpression measured by IHC, FISH, or real-time PCR was found in 5.0%, 5.0%, and 22.5%, respectively. HER2 negative results measured by IHC, FISH, or real-time PCR were found in 95%, 92.5%, and 60.0%, respectively. Analysis failed for IHC, FISH, or real-time PCR in 0%, 2.5%, or 17.5% of cases. Concordance between IHC and FISH, IHC and real-time PCR, or FISH and real-time PCR were 89.7%, 72.7%, or 78.1%, respectively. Only few ovarian cancer patients were HER2 overexpressed measured by IHC or FISH and thus could be eligible for antibody-based therapy with trastuzumab (Herceptin). Interestingly, we find an increased number of HER2 positive patients by real-time PCR analysis on microdissected cancer cells, suggesting a number of HER2 positive patients not detected by current methods. Thus, the concept of quantitative measurement of HER2 on microdissected cancer cells should be explored further.
Collapse
Affiliation(s)
- Thore Hillig
- Department of Clinical Biochemistry, Hillerød Hospital University of Copenhagen, 3400 Hillerød, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Xie Y, Long Q, Wu Q, Shi S, Dai M, Liu Y, Liu L, Gong C, Qian Z, Wei Y, Zhao X. Improving therapeutic effect in ovarian peritoneal carcinomatosis with honokiol nanoparticles in a thermosensitive hydrogel composite. RSC Adv 2012. [DOI: 10.1039/c2ra20612a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
16
|
Fischer E, Grünberg J, Cohrs S, Hohn A, Waldner-Knogler K, Jeger S, Zimmermann K, Novak-Hofer I, Schibli R. L1-CAM-targeted antibody therapy and (177)Lu-radioimmunotherapy of disseminated ovarian cancer. Int J Cancer 2011; 130:2715-21. [PMID: 21796623 DOI: 10.1002/ijc.26321] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/15/2011] [Accepted: 07/01/2011] [Indexed: 11/07/2022]
Abstract
The L1-cell adhesion molecule (L1-CAM) is highly expressed in various cancer types including ovarian carcinoma but is absent from most normal tissue. A chimeric monoclonal antibody, chCE7, specifically binds to human L1-CAM and exhibits anti-proliferative effects on L1-CAM-expressing tumor cells. The goal of this study was to evaluate the efficacy of a novel (177)Lu-chCE7 radioimmunotherapeutic agent and to compare it to a treatment protocol with unlabeled, growth-inhibiting chCE7 in a mouse xenograft model of disseminated ovarian cancer. chCE7agl, an aglycosylated IgG1 variant with improved pharmacokinetics, was conjugated with 1,4,7,10-tetraazacyclododecane-N-N'-N'-N‴-tetraacetic acid (DOTA) and labeled with the low-energy β-emitter (177)Lu. Tumor growth and survival were assessed after a single i.v. dose of 8 MBq (60 μg) radioimmunoconjugate in nude mice bearing either subcutaneous or intraperitoneal SKOV3.ip1 human ovarian cancer tumors. Therapeutic efficacy was compared with three times weekly i.p. administration of 10 mg/kg unconjugated chCE7. In vivo analysis of (177)Lu-chCE7agl biodistribution demonstrated high and specific accumulation of radioactivity at the tumor site with maximal tumor uptake of up to 48.0 ± 8.1% ID/g at 168 h postinjection. A single treatment with (177)Lu-DOTA-chCE7agl caused significant retardation of tumor growth and prolonged median survival from 33 to 71 days, while administration of a nontargeted (177)Lu-immunoconjugate had no beneficial effect. Three times weekly i.p. application of unlabeled chCE7 10 mg/kg similarly increased survival from 44 to 72 days. We conclude that a single dose of (177)Lu-DOTA-chCE7agl is as effective as repeated administration of nonradioactive chCE7 for treatment of small intraperitoneal tumors expressing L1-CAM.
Collapse
Affiliation(s)
- Eliane Fischer
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Xiang X, Feng M, Felder M, Connor JP, Man YG, Patankar MS, Ho M. HN125: A Novel Immunoadhesin Targeting MUC16 with Potential for Cancer Therapy. J Cancer 2011; 2:280-91. [PMID: 21611109 PMCID: PMC3100680 DOI: 10.7150/jca.2.280] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 05/16/2011] [Indexed: 02/06/2023] Open
Abstract
Background: The mucin MUC16 expresses the repeating peptide epitope CA125 that has been known for decades to be a well-validated cancer marker that is overexpressed on the cell surface of ovarian cancers and other malignant tumors. In spite of recent efforts to make mouse monoclonal antibodies to MUC16 to treat ovarian cancer, a human monoclonal antibody against this mucin has not been described. MUC16 interacts with mesothelin, a protein that mediates heterotypic cancer cell adhesion, indicating that MUC16 and mesothelin play an important role in the peritoneal implantation and metastasis of ovarian tumors. Therefore, a suitable candidate for therapeutic targeting of MUC16 would functionally block the interaction of MUC16 and mesothelin. Methodology/Principal Findings: Here we report the generation of a novel immunoadhesin, HN125, against MUC16 that consists of a functional MUC16 binding domain of mesothelin (IAB) and the Fc portion of a human antibody IgG1. The yield for purified HN125 proteins is over 100 µg/mL of HEK-293 culture supernatant. We show that HN125 has high and specific affinity for MUC16-expressing cancer cells by flow cytometry and immunohistochemistry. HN125 has the ability to disrupt the heterotypic cancer cell adhesion mediated by the MUC16-mesothelin interaction. Moreover, it elicits strong antibody-dependent cell mediated cytotoxicity against MUC16-positive cancer cells in vitro. Conclusion/Significance: This report describes a novel human immunotherapeutic agent highly specific for MUC16 with potential for treating ovarian cancer and other MUC16-expressing tumors. Because of its lower immunogenicity in patients, a fully human protein is the most desirable format for clinical applications. We believe that the methods developed here may apply to the generation of other tumor-targeting immunoadhesins when it is difficult to obtain a human monoclonal antibody to a given antigen for clinical applications. The resultant immunoadhesins can have advantages usually found in monoclonal antibodies such as ease of purification, high binding affinity and effector functions.
Collapse
Affiliation(s)
- Xinran Xiang
- 1. Antibody Therapy Unit, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Jakobsen JN, Sørensen JB. Review on clinical trials of targeted treatments in malignant mesothelioma. Cancer Chemother Pharmacol 2011; 68:1-15. [PMID: 21553148 DOI: 10.1007/s00280-011-1655-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 04/14/2011] [Indexed: 12/21/2022]
Abstract
PURPOSE Malignant mesothelioma (MM) is an aggressive tumor of the serosal surfaces with a poor prognosis. Advances in the understanding of tumor biology have led to the development of several targeted treatments, which have been evaluated in clinical trials. This article is a comprehensive review of all clinical trials evaluating the effect of targeted treatments in MM. METHODS An extensive literature search was performed in January 2011 using pubmed and medline. No constraints on publication date were applied. RESULTS Thirty-two trials exploring 17 different targeted agents in MM were found. Treatment in first- and second-line targeted agents induced response rates ranging from 0-14% and 0-16%, respectively. The tyrosine kinase inhibitor sunitinib induced partial response in 10% and stable disease in 66% of MPM patients as second-line treatment. A preliminary analysis of a phase II/III trial suggests that addition of bevacizumab to pemetrexed and cisplatin first-line treatment significantly improves disease control (CR + PR + SD) in the bevacizumab arm (73.5%) compared with treatment with pemetrexed and cisplatin without bevacizumab (43.2%) (P = 0.010). Another phase II trial did not observe any significant clinical benefit of adding of bevacizumab to gemcitabine and cisplatin. CONCLUSIONS Disease stabilization is reported in some patients with several targeted treatments and might be beneficial in subgroups of patients or in combination with classic chemotherapy. None of the hitherto explored targeted treatments can currently be recommended as standard treatment in MM.
Collapse
|
19
|
Wållberg H, Orlova A, Altai M, Hosseinimehr SJ, Widström C, Malmberg J, Ståhl S, Tolmachev V. Molecular design and optimization of 99mTc-labeled recombinant affibody molecules improves their biodistribution and imaging properties. J Nucl Med 2011; 52:461-9. [PMID: 21321280 DOI: 10.2967/jnumed.110.083592] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED Affibody molecules are a recently developed class of targeting proteins based on a nonimmunoglobulin scaffold. The small size (7 kDa) and subnanomolar affinity of Affibody molecules enables high-contrast imaging of tumor-associated molecular targets, particularly human epidermal growth factor receptor type 2 (HER2). (99m)Tc as a label offers advantages in clinical practice, and earlier studies demonstrated that (99m)Tc-labeled recombinant Affibody molecules with a C-terminal cysteine could be used for HER2 imaging. However, the renal retention of radioactivity exceeded tumor uptake, which might complicate imaging of metastases in the lumbar region. The aim of this study was to develop an agent with low renal uptake and preserved tumor targeting. METHODS A series of recombinant derivatives of the HER2-binding Z(HER2)(:342) Affibody molecule with a C-terminal chelating sequence, -GXXC (X denoting glycine, serine, lysine, or glutamate), was designed. The constructs were labeled with (99m)Tc and evaluated in vitro and in vivo. RESULTS All variants were stably labeled with (99m)Tc, with preserved capacity to bind specifically to HER2-expressing cells in vitro and in vivo. The composition of the chelating sequence had a clear influence on the cellular processing and biodistribution properties of the Affibody molecules. The best variant, (99m)Tc-Z(HER2)(:V2), with the C-terminal chelating sequence -GGGC, provided the lowest radioactivity retention in all normal organs and tissues including the kidneys. (99m)Tc-Z(HER2)(:V2) displayed high uptake of radioactivity in HER2-expressing xenografts, 22.6 ± 4.0 and 7.7 ± 1.5 percentage injected activity per gram of tissue at 4 h after injection in SKOV-3 (high HER2 expression) and DU-145 (low HER2 expression) tumors, respectively. In both models, the tumor uptake exceeded the renal uptake. CONCLUSION These results demonstrate that the biodistribution properties of recombinant (99m)Tc-labeled Affibody molecules can be optimized by modification of the C-terminal cysteine-containing chelating sequence. (99m)Tc-Z(HER2)(:V2) is a promising candidate for further development as a diagnostic radiopharmaceutical for imaging of HER2-expressing tumors. These results may be useful for the development of imaging agents based on other Affibody molecules and, hopefully, other scaffolds.
Collapse
Affiliation(s)
- Helena Wållberg
- School of Biotechnology, AlbaNova University Center, Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Van Elssen CHMJ, Frings PWH, Bot FJ, Van de Vijver KK, Huls MB, Meek B, Hupperets P, Germeraad WTV, Bos GMJ. Expression of aberrantly glycosylated Mucin-1 in ovarian cancer. Histopathology 2011; 57:597-606. [PMID: 20955385 DOI: 10.1111/j.1365-2559.2010.03667.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIMS Mucin 1 (MUC1) is an important tumour-associated antigen (TAA), both overexpressed and aberrantly glycosylated in adenocarcinomas. The aim of this study was to examine the MUC1-glycosylation status of primary ovarian adenocarcinomas and metastatic lesions. METHODS AND RESULTS Paraffin-embedded tissue sections of 37 primary ovarian adenocarcinomas representing all histotypes (22 serous, five mucinous, two clear-cell, eight endometrioid), four serous borderline tumours with intraepithelial carcinoma, seven sections of ovarian endometriosis and 13 metastatic lesions were analysed by immunohistochemistry. Non-neoplastic ovarian surface epithelium and serous cystadenomas were used as controls. All epithelia expressed MUC1 protein. Of primary tumours, 76% expressed the differentiation-dependent glycoform and 84% the cancer-associated glycoform (Tn/Sialyl-Tn-epitopes). In metastatic lesions this was 77% and 85%, respectively. Notably, in 57% of ovarian endometriosis and 75% of intraepithelial lesions, the cancer-associated MUC1 epitopes were expressed, whereas normal ovarian surface epithelium and serous cystadenomas did not express these epitopes. CONCLUSIONS The underglycosylated MUC1 epitopes are expressed by all histotypes of primary ovarian adenocarcinomas, by the vast majority of metastatic lesions and by possible ovarian cancer precursor lesions, but not by normal ovarian tissue. These results indicate that MUC1-associated Tn/STn-epitopes are important targets for immunotherapy and diagnostic imaging in ovarian cancer patients.
Collapse
Affiliation(s)
- Catharina H M J Van Elssen
- Department of Internal Medicine, Division of Haematology, Maastricht University Medical Center, Maastricht, the Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kenny HA, Leonhardt P, Ladanyi A, Yamada SD, Montag A, Im HK, Jagadeeswaran S, Shaw DE, Mazar AP, Lengyel E. Targeting the urokinase plasminogen activator receptor inhibits ovarian cancer metastasis. Clin Cancer Res 2011; 17:459-71. [PMID: 21149615 PMCID: PMC3073583 DOI: 10.1158/1078-0432.ccr-10-2258] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To understand the functional and preclinical efficacy of targeting the urokinase plasminogen activator receptor (u-PAR) in ovarian cancer. EXPERIMENTAL DESIGN Expression of u-PAR was studied in 162 epithelial ovarian cancers, including 77 pairs of corresponding primary and metastatic tumors. The effect of an antibody against u-PAR (ATN-658) on proliferation, adhesion, invasion, apoptosis, and migration was assessed in 3 (SKOV3ip1, HeyA8, and CaOV3) ovarian cancer cell lines. The impact of the u-PAR antibody on tumor weight, number, and survival was examined in corresponding ovarian cancer xenograft models and the mechanism by which ATN-658 blocks metastasis was explored. RESULTS Only 8% of all ovarian tumors were negative for u-PAR expression. Treatment of SKOV3ip1, HeyA8, and CaOV3 ovarian cancer cell lines with the u-PAR antibody inhibited cell invasion, migration, and adhesion. In vivo, anti-u-PAR treatment reduced the number of tumors and tumor weight in CaOV3 and SKOV3ip1 xenografts and reduced tumor weight and increased survival in HeyA8 xenografts. Immunostaining of CaOV3 xenograft tumors and ovarian cancer cell lines showed an increase in active-caspase 3 and TUNEL staining. Treatment with u-PAR antibody inhibited α(5)-integrin and u-PAR colocalization on primary human omental extracellular matrix. Anti-u-PAR treatment also decreased the expression of urokinase, u-PAR, β(3)-integrin, and fibroblast growth factor receptor-1 both in vitro and in vivo. CONCLUSIONS This study shows that an antibody against u-PAR reduces metastasis, induces apoptosis, and reduces the interaction between u-PAR and α(5)-integrin. This provides a rationale for targeting the u-PAR pathway in patients with ovarian cancer and for further testing of ATN-658 in this indication.
Collapse
Affiliation(s)
- Hilary A. Kenny
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology – Center for Integrative Science, University of Chicago, Chicago, IL, 60637, USA
| | - Payton Leonhardt
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology – Center for Integrative Science, University of Chicago, Chicago, IL, 60637, USA
| | - Andras Ladanyi
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology – Center for Integrative Science, University of Chicago, Chicago, IL, 60637, USA
| | - S. Diane Yamada
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology – Center for Integrative Science, University of Chicago, Chicago, IL, 60637, USA
| | - Anthony Montag
- Department of Pathology, University of Chicago, Chicago, IL, 60637, USA
| | - Hae Kyung Im
- Department of Health Studies, University of Chicago, Chicago, IL, 60637, USA
| | - Sujatha Jagadeeswaran
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology – Center for Integrative Science, University of Chicago, Chicago, IL, 60637, USA
| | - David E. Shaw
- D. E. Shaw Research and Center for Computational Biology and Bioinformatics, Columbia University, New York, NY, 10032, USA
| | - Andrew P. Mazar
- Chemistry of Life Processes Institute and Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL 60208, USA
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology – Center for Integrative Science, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
22
|
Targeted microbeads for attraction and induction of specific innate immune response in the tumor microenvironment. Vaccine 2010; 28:7279-87. [PMID: 20831916 DOI: 10.1016/j.vaccine.2010.08.083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 08/16/2010] [Accepted: 08/22/2010] [Indexed: 11/21/2022]
Abstract
Antitumor activity of molecules and cells of the innate immune system has been reported. Here we propose a method for targeting preferred innate immune cells and magnifying their tumoricidal effect at the tumor microenvironment, by modular multiple-component complexes (termed TILTAN). As a model, micro-scale complexes were assembled carrying monoclonal anti-HER2 antibodies, lipopolysaccharide and/or mannose. The complexes showed high binding capacity to HER2-positive cancer cells in vitro, high induction of interleukin-1 RNA transcription by the activated monocytes and ability to mediate monocytes' attachment to HER2-positive cells. TILTAN treatment was found safe in in vivo testing and induced change in interleukin-1 RNA transcription in tumors xenografts. We thus present a new vision of targeting a desired innate immune response to the tumor microenvironment.
Collapse
|
23
|
Abstract
Over the past decade, a multitude of targeted agents have been explored in the treatment of advanced non-small cell lung cancer (NSCLC). Thus far, two broad classes of agents have been implemented in clinical practice: (a) vascular endothelial growth factor (VEGF)-directed therapies and (b) antagonists of the epidermal growth factor receptor (EGFR). In the former category, the agent bevacizumab (a monoclonal antibody) has shown landmark improvements in survival when added to cytotoxic therapy. Small molecule tyrosine kinase inhibitors (TKI) targeting the VEGF receptor (i.e., sunitinib, sorafenib, and vandetanib) show activity in phase II clinical studies. With respect to EGFR-directed therapies, the TKIs gefitinib and erlotinib have shown significant benefit, and have uncovered valuable information about the biology of lung cancer. Outside of therapies directed specifically at VEGF- and EGFR-mediated signaling, trials evaluating insulin-like growth factor-1 receptor (IGF-IR)-targeting agents, cyclooxygenase-2 (COX-2) inhibitors, c-met inhibitors, irreversible pan-HER inhibitors, mammalian target of rapamycin (mTOR) inhibitors, and histone deacetylase (HDAC) inhibitors are ongoing. Inhibitors of ALK show great promise in patients with the relevant gene translocation. Herein, the clinical development of novel therapies for NSCLC is described, including some discussion of relevant biomarkers and determination of synergy with both cytotoxic therapy and other targeted agents.
Collapse
Affiliation(s)
- Sumanta Kumar Pal
- Division of Genitourinary Malignancies, Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Phone: (626) 256-4673 Fax: (626) 301-8233
| | - Robert A. Figlin
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Phone: (626) 256-4673, Fax: (626) 301-8233
| | - Karen Reckamp
- Division of Thoracic Malignancies, Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Phone: (626) 256-4673
| |
Collapse
|
24
|
Cubillos-Ruiz JR, Fiering S, Conejo-Garcia JR. Nanomolecular targeting of dendritic cells for ovarian cancer therapy. Future Oncol 2010; 5:1189-92. [PMID: 19852731 DOI: 10.2217/fon.09.101] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
25
|
Antivascular therapy for epithelial ovarian cancer. JOURNAL OF ONCOLOGY 2009; 2010:372547. [PMID: 20072701 PMCID: PMC2804796 DOI: 10.1155/2010/372547] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Accepted: 09/28/2009] [Indexed: 12/19/2022]
Abstract
Ovarian cancer is the fifth largest cancer killer in women. Improved understanding of the molecular pathways implicated in the pathogenesis of ovarian cancer has led to the investigation of novel targeted therapies. Ovarian cancer is characterized by an imbalance between pro- and antiangiogenic factors in favor of angiogenesis activation. Various antivascular strategies are currently under investigation in ovarian cancer. They can schematically be divided into antiangiogenic and vascular-disrupting therapies. This paper provides a comprehensive review of these new treatments targeting the tumor vasculature in this disease. Promising activities have been detected in phase II trials, and results of phase III clinical trials are awaited eagerly.
Collapse
|
26
|
Histone deacetylase inhibitor therapy in epithelial ovarian cancer. JOURNAL OF ONCOLOGY 2009; 2010:458431. [PMID: 20052384 PMCID: PMC2801002 DOI: 10.1155/2010/458431] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 09/06/2009] [Accepted: 10/13/2009] [Indexed: 11/26/2022]
Abstract
Since epigenetic alterations are believed to be involved in the repression of tumor suppressor genes and promotion of tumorigenesis in ovarian cancers, novel compounds endowed with a histone deacetylase (HDAC) inhibitory activity are an attractive therapeutic approach. In this review, we discuss the biologic and therapeutic effects of HDAC inhibitors (HDACIs) in treating ovarian cancer. HDACIs were able to mediate inhibition of cell growth, cell cycle arrest, apoptosis, and expression of genes related to the malignant phenotype in a variety of ovarian cancer cell lines. Furthermore, HDACIs were able to induce the accumulation of acetylated histones in the chromatin of the p21WAF1 gene in human ovarian carcinoma cells. In xenograft models, some of HDACIs have demonstrated antitumor activity with only few side effects. Some clinical trials demonstrate that HDACI drugs provide an important class of new mechanism-based therapeutics for ovarian cancer. In this review, we discuss the biologic and therapeutic effects of HDACIs in treating ovarian cancer, especially focusing on preclinical studies and clinical trials.
Collapse
|