Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Int J Gynecol Cancer ; 33(9): 1458-1463, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37666539

ABSTRACT

BACKGROUND: Treatment options for patients with platinum-resistant/refractory ovarian cancers are limited and only marginally effective. The development of novel, more effective therapies addresses a critical unmet medical need. Olvimulogene nanivacirepvec (Olvi-Vec), with its strong immune modulating effect on the tumor microenvironment, may provide re-sensitization to platinum and clinically reverse platinum resistance or refractoriness in platinum-resistant/refractory ovarian cancer. PRIMARY OBJECTIVE: The primary objective is to evaluate the efficacy of intra-peritoneal Olvi-Vec followed by platinum-based chemotherapy and bevacizumab in patients with platinum-resistant/refractory ovarian cancer. STUDY HYPOTHESIS: This phase III study investigates Olvi-Vec oncolytic immunotherapy followed by platinum-based chemotherapy and bevacizumab as an immunochemotherapy evaluating the hypothesis that such sequential combination therapy will prolong progression-free survival (PFS) and bring other clinical benefits compared with treatment with platinum-based chemotherapy and bevacizumab. TRIAL DESIGN: This is a multicenter, prospective, randomized, and active-controlled phase III trial. Patients will be randomized 2:1 into the experimental arm treated with Olvi-Vec followed by platinum-doublet chemotherapy and bevacizumab or the control arm treated with platinum-doublet chemotherapy and bevacizumab. MAJOR INCLUSION/EXCLUSION CRITERIA: Eligible patients must have recurrent, platinum-resistant/refractory, non-resectable high-grade serous, endometrioid, or clear-cell ovarian, fallopian tube, or primary peritoneal cancer. Patients must have had ≥3 lines of prior chemotherapy. PRIMARY ENDPOINT: The primary endpoint is PFS in the intention-to-treat population. SAMPLE SIZE: Approximately 186 patients (approximately 124 patients randomized to the experimental arm and 62 to the control arm) will be enrolled to capture 127 PFS events. ESTIMATED DATES FOR COMPLETING ACCRUAL AND PRESENTING RESULTS: Expected complete accrual in 2024 with presentation of primary endpoint results in 2025. TRIAL REGISTRATION: NCT05281471.


Subject(s)
Ovarian Neoplasms , Viral Vaccines , Humans , Female , Bevacizumab , Prospective Studies , Carcinoma, Ovarian Epithelial , Platinum , Ovarian Neoplasms/drug therapy , Tumor Microenvironment
2.
J Ovarian Res ; 15(1): 120, 2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36324187

ABSTRACT

A leading theory for ovarian carcinogenesis proposes that inflammation associated with incessant ovulation is a driver of oncogenesis. Consistent with this theory, nonsteroidal anti-inflammatory drugs (NSAIDs) exert promising chemopreventive activity for ovarian cancer. Unfortunately, toxicity is associated with long-term use of NSAIDs due to their cyclooxygenase (COX) inhibitory activity. Previous studies suggest the antineoplastic activity of NSAIDs is COX independent, and rather may be exerted through phosphodiesterase (PDE) inhibition. PDEs represent a unique chemopreventive target for ovarian cancer given that ovulation is regulated by cyclic nucleotide signaling. Here we evaluate PDE10A as a novel therapeutic target for ovarian cancer. Analysis of The Cancer Genome Atlas (TCGA) ovarian tumors revealed PDE10A overexpression was associated with significantly worse overall survival for patients. PDE10A expression also positively correlated with the upregulation of oncogenic and inflammatory signaling pathways. Using small molecule inhibitors, Pf-2545920 and a novel NSAID-derived PDE10A inhibitor, MCI-030, we show that PDE10A inhibition leads to decreased ovarian cancer cell growth and induces cell cycle arrest and apoptosis. We demonstrate these pro-apoptotic properties occur through PKA and PKG signaling by using specific inhibitors to block their activity. PDE10A genetic knockout in ovarian cancer cells through CRISP/Cas9 editing lead to decreased cell proliferation, colony formation, migration and invasion, and in vivo tumor growth. We also demonstrate that PDE10A inhibition leads to decreased Wnt-induced ß-catenin nuclear translocation, as well as decreased EGF-mediated activation of RAS/MAPK and AKT pathways in ovarian cancer cells. These findings implicate PDE10A as novel target for ovarian cancer chemoprevention and treatment.


Subject(s)
Ovarian Neoplasms , beta Catenin , Female , Humans , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , beta Catenin/genetics , beta Catenin/metabolism , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , ras Proteins/metabolism
3.
Int J Gynecol Cancer ; 31(11): 1403-1407, 2021 11.
Article in English | MEDLINE | ID: mdl-34088749

ABSTRACT

OBJECTIVE: To describe the participation of minority women in clinical trials using immunologic agents for breast and gynecologic cancers. METHODS: A retrospective review of completed clinical trials involving immunotherapy for breast and gynecologic cancers was performed. Completed trials were examined for data on race, tumor type, and start year. Minority enrollment was stratified by tumor site. Based on Center for Disease Control and Prevention age-adjusted incidence for race, expected and observed ratios of racial participation were calculated and compared using Χ2 testing, p≤0.05. RESULTS: A total of 53 completed immunotherapy clinical trials involving 8820 patients were reviewed. Breast cancer trials were most common (n=24) and involved the most patients (n=6248, 71%). Racial breakdown was provided in 41 studies (77%) for a total of 7201 patients. Race reporting was lowest in uterine (n=4, 67%) and cervical cancer trials (n=6, 67%), and highest in ovarian cancer trials (n=12, 86%). White patients comprised 70% (n=5022) of all the patients included. Only 5% of patients involved were black (n=339), and 83% of these patients (n=282) were enrolled in breast cancer trials. Observed enrollment of black women was 32-fold lower for ovarian, 19-fold lower for cervical, 15-fold lower for uterine, and 11-fold lower for breast cancer than expected. While all trials reported race between 2013 and 2015, no consistent trend was seen towards increasing race reporting or in enrollment of black patients over time. CONCLUSION: Racial disparities exist in clinical trials evaluating immunologic agents for breast and gynecologic cancers. Recruitment of black women is particularly low. In order to address inequity in outcomes for these cancers, it is crucial that significant attention be directed towards minority representation in immuno-oncologic clinical trials.


Subject(s)
Breast Neoplasms/ethnology , Clinical Trials as Topic/statistics & numerical data , Genital Neoplasms, Female/ethnology , Health Status Disparities , Black or African American/statistics & numerical data , Breast Neoplasms/immunology , Female , Genital Neoplasms, Female/immunology , Hispanic or Latino/statistics & numerical data , Humans , Immunotherapy , Patient Selection , Retrospective Studies , White People/statistics & numerical data
4.
Cancer ; 126(22): 4948-4956, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32910478

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors are being considered for locally advanced cervical cancer (LACC) together with standard-of-care pelvic chemoradiation (CRT). However, the safety of the combination and its optimal schedule are unknown. Defining the safety of the combination is a primary objective of a study examining concurrent and sequential schedules. This article presents a safety analysis that was fully accrued and met reporting requirements. METHODS: Pembrolizumab was given after CRT (arm 1) or during CRT (arm 2) according to a randomized phase 2 design. Patients who were 18 years old or older and had LACC (stages IB-IVA according to the 2009 International Federation of Gynecology and Obstetrics system) were randomized 1:1 to the treatment regimens. The CRT was identical in the 2 arms. Pembrolizumab was administered every 3 weeks for 3 doses; no maintenance was allowed. All patients receiving any treatment were evaluated for safety. Safety assessments included the incidence and severity of adverse events (AEs) and the occurrence of protocol-defined dose-limiting toxicity (DLT) through 30 days after the last pembrolizumab infusion. RESULTS: As of August 2019, 52 of the 88 planned patients had completed treatment and were evaluable for toxicity. Treatment-related grade 2 or higher toxicity was experienced by 88%; 11 had at least 1 grade 4 AE, and another 23 had at least 1 grade 3 AE. Grade 1 or higher diarrhea was reported in 34 patients (65%; 50% of these were grade 1), and there was no difference between arms (63% in arm 1 vs 68% in arm 2). Two patients experienced 3 DLTs. Most patients completed cisplatin (100% in arm 1 vs 82% in arm 2); 83% in both arms completed all pembrolizumab. CONCLUSIONS: Preliminary results support the safety and feasibility of adding pembrolizumab to pelvic CRT concurrently or sequentially. LAY SUMMARY: Pembrolizumab is a humanized antibody against programmed cell death protein 1 that is used in cancer immunotherapy. Preliminary data suggest that pembrolizumab can be safely combined with chemotherapy and pelvic radiation in the treatment of locally advanced cervical cancer. Future studies of the addition of immunotherapy to traditional chemoradiation are planned to determine the best way to deliver the treatment and whether any improvement is seen with the addition of immunotherapy to traditional therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Pelvis/pathology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/radiotherapy , Antibodies, Monoclonal, Humanized/pharmacology , Female , Humans , Male
5.
Cancer ; 123(6): 977-984, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28263384

ABSTRACT

BACKGROUND: Mesothelium vascular cell adhesion molecule-1 (VCAM-1) expression in the metastatic epithelial ovarian cancer (EOC) microenvironment is induced by tumor and mediates tumor cell invasion. VCAM-1 imaging suggests expression during treatment is an indicator of platinum resistance. Here, we assess the potential prognostic significance of mesothelium VCAM-1 expression and prospectively evaluate whether soluble VCAM-1 (sVCAM-1) is a surrogate for mesothelium expression. METHODS: A retrospective review of EOC patients was performed to evaluate outcomes with mesothelium VCAM-1 expression determined by immunohistochemistry of peritoneum or omentum specimens. A prospective cohort of EOC patients was identified and followed through primary treatment. Serum for sVCAM-1 evaluation, which was performed via enzyme-linked immunosorbent assay, was collected before surgery or neoadjuvant chemotherapy and at each treatment cycle. Peritoneal specimens were obtained during debulking to assess mesothelial VCAM-1 expression. RESULTS: A retrospective review identified 54 advanced-stage EOC patients. Patients expressing mesothelium VCAM-1 had shortened overall survival (44 vs 79 months, P = 0.035) and progression-free survival (18 vs 67 months, P = 0.010); the median time to platinum resistance was 36 months for VCAM-1-expressing patients and not yet determined for the VCAM-1-negative group. In our prospective observational cohort, 18 EOC patients completed primary treatment; 3 were negative for mesothelium VCAM-1 expression, and sVCAM-1 did not vary between groups. CONCLUSIONS: Mesothelium VCAM-1 expression is negatively associated with progression-free and overall survival in EOC. This is especially compelling in light of previous data suggesting that persistent VCAM-1 expression during treatment is an indicator of platinum resistance. Our pilot study had insufficient cases to determine whether sVCAM-1 would substitute for mesothelium expression. Cancer 2017;123:977-84. © 2016 American Cancer Society.


Subject(s)
Epithelium/metabolism , Gene Expression , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Vascular Cell Adhesion Molecule-1/genetics , Adult , Aged , Biomarkers, Tumor , Carcinoma, Ovarian Epithelial , Combined Modality Therapy , Epithelium/pathology , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasms, Glandular and Epithelial/diagnosis , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/therapy , Prognosis , Retrospective Studies , Treatment Outcome , Vascular Cell Adhesion Molecule-1/blood
6.
Gynecol Oncol ; 145(2): 329-333, 2017 05.
Article in English | MEDLINE | ID: mdl-28215839

ABSTRACT

BACKGROUND: Our objective was to evaluate racial treatment and survival disparities in black women with ovarian cancer in the Deep South and to determine how environmental factors / socioeconomic status (SES) influence survival. METHODS: A retrospective study of ovarian cancer patients from 2007 to 2014 was performed. Socioeconomic status (SES) was obtained though U.S. Census block data and compared using Yost scores. Comparisons were performed using standard statistical approaches. RESULTS: A total of 393 patients were evaluated, 325 (83%) white and 68 (17%) black. Demographic information and surgical approach were similar in each racial group. However, compared to whites, black patients had lower rates of optimal debulking [89% vs. 71%, respectively (p=0.001)] and intraperitoneal chemotherapy (19% vs. 11%, p=0.01). Black women had lower SES parameters including education, income, and poverty. As a result, more black patients had the lowest SES (SES-1) when compared to white patients (17% vs. 41%, p<0.001). When controlling for these factors by cox regression analysis, a survival disadvantage was seen in black women for both progression free survival (16 vs. 27months, p=0.003) and overall survival (42 vs. 88months, p<0.001). CONCLUSIONS: Despite controlling for clinical and environmental factors, a survival disadvantage was still observed in black patients with ovarian cancer in the Deep South. Black women had lower optimal debulking rates and more platinum resistant disease. These data suggest other factors like tumor biology may play a role in racial survival differences, however, more research is needed to determine this causation.


Subject(s)
Black People/statistics & numerical data , Health Status Disparities , Neoplasms, Glandular and Epithelial/ethnology , Ovarian Neoplasms/ethnology , Alabama/epidemiology , Carcinoma, Ovarian Epithelial , Cohort Studies , Disease-Free Survival , Female , Humans , Middle Aged , Neoplasm Staging , Neoplasms, Glandular and Epithelial/epidemiology , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/mortality , Retrospective Studies , Socioeconomic Factors , White People/statistics & numerical data
7.
Gynecol Oncol ; 132(2): 455-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24378876

ABSTRACT

OBJECTIVE: The inability to successfully treat women with ovarian cancer is due to the presence of metastatic disease at diagnosis and the development of platinum resistance. Ovarian cancer metastasizes throughout the peritoneal cavity by attaching to and invading through the mesothelium lining the peritoneum using a mechanism that involves α4ß1 integrin and its ligand (vascular cell adhesion molecule) VCAM-1. Integrin α4ß1 expression on tumor cells is known to confer protection from therapy in other cancers, notably multiple myeloma. We evaluated the role of α4ß1 integrin in response to platinum-based therapy in a mouse model of peritoneal ovarian cancer metastasis by treatment with a humanized anti-α4ß1 integrin function-blocking antibody. METHODS: Integrin α4ß1 expression on primary human ovarian cancer cells, fallopian tube and ovarian surface epithelia and fresh tumor was assessed by flow-cytometry. The therapeutic impact of anti-α4ß1 treatment was assessed in murine models of platinum-resistant peritoneal disease and in vitro using the platinum resistant ovarian cancer cell lines. RESULTS: Treatment of tumor-bearing mice with human-specific α4ß1 integrin function-blocking antibodies, anti-VCAM-1 antibody or carboplatin alone had no effect on tumor burden compared to the IgG control group. However, the combined treatment of anti-α4ß1 integrin or anti-VCAM-1 with carboplatin significantly reduced tumor burden. In vitro, the combination of carboplatin and anti-α4ß1 integrin antibodies resulted in increased cell death and doubling time. CONCLUSIONS: Our findings support a role for α4ß1 integrin in regulating treatment response to carboplatin, implicating α4ß1 integrin as a potential therapeutic target to influence platinum responsiveness in otherwise resistant disease.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carboplatin/pharmacology , Integrin alpha4beta1/antagonists & inhibitors , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Carboplatin/administration & dosage , Carcinoma, Ovarian Epithelial , Cell Growth Processes/drug effects , Cell Line, Tumor , Drug Synergism , Female , Humans , Integrin alpha4beta1/biosynthesis , Integrin alpha4beta1/immunology , Integrin alpha4beta1/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Mice , Mice, Nude , Natalizumab , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Rats , Xenograft Model Antitumor Assays
8.
Int J Gynecol Cancer ; 24(1): 91-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24362716

ABSTRACT

OBJECTIVE: Literature suggests that para-aortic lymphadenectomy (para-aortic lymph node dissection [PALND]) has a therapeutic benefit for women with intermediate- to high-risk endometrial adenocarcinoma. We hypothesized that the observed survival advantage of PALND is a reflection of the general health of the patient rather than a therapeutic benefit of surgery. METHODS: Women with intermediate- to high-risk endometrial adenocarcinoma diagnosed from 2002 to 2009 at a single institution were identified. Medical comorbidities, pathology, and survival information were abstracted from the medical record. The χ test or the t test was used for univariate analysis. Overall survival (OS) and disease-specific survival (DSS) were calculated using the Kaplan-Meier method. RESULTS: A total of 253 women with a mean age of 64 years were identified. Of these women, 174 had a pelvic lymphadenectomy (pelvic lymph node dissection [PLND]) and 82 had PLND and PALND. The rate of positive nodes was 13% (23/174) for the women who had PLND and was 7% (6/82) for those who had PLND and PALND. Only 1.2% (1/82) of the women who had PLND and PALND had negative pelvic but positive para-aortic nodes. The patients who had PALND had a lower body mass index and were less likely to have significant medical comorbidities. The patients who had PALND had improved 5-year OS (96% vs 82%, P = 0.007) but no difference in 5-year DSS (96% vs 89%, P value = not significant). CONCLUSIONS: Women with intermediate- to high-risk endometrial adenocarcinoma who undergo PALND have improved OS but no improvement in DSS. The lack of difference in DSS supports the hypothesis that underlying comorbidities as opposed to lack of PALND result in poorer outcome.


Subject(s)
Adenocarcinoma/surgery , Endometrial Neoplasms/surgery , Lymph Node Excision , Registries , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Endometrial Neoplasms/mortality , Female , Humans , Middle Aged , Virginia/epidemiology
9.
J Nucl Med ; 54(11): 1883-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24029657

ABSTRACT

UNLABELLED: The inability to successfully treat women with ovarian cancer is due in large part to the advanced stage of disease at diagnosis, the development of platinum resistance, and the lack of sensitive methods to monitor tumor progression and response to treatment. Vascular cell adhesion molecule-1 (VCAM-1) is expressed on the mesothelium of ovarian cancer patients. We investigated VCAM-1 expression as a marker of peritoneal metastasis and tumor response to platinum-based chemotherapy. METHODS: Peritoneal or omental biopsies obtained from women diagnosed with stage I, stage II, or stage III/IV ovarian cancer were evaluated by immunohistochemistry. The effects of carboplatin on mesothelial VCAM-1 expression were determined in cultured cells by Western blot. Radiolabeled VCAM-1-specific peptide imaging probes and SPECT were used in a mouse model of ovarian cancer peritoneal metastasis to identify VCAM-1 as a viable imaging target. RESULTS: VCAM-1 expression correlated with tumor stage. All specimens from stage I patients were negative, whereas 29% of stage II patients and 73% of stage III/IV patients were positive. Although most women with advanced stage disease expressed VCAM-1, the incidence of expression was reduced among women who received neoadjuvant chemotherapy, suggesting a role for chemotherapy in regulating VCAM-1 expression. Treatment of mesothelial cells in culture with carboplatin resulted in a transient decrease in VCAM-1 expression 4 h after treatment that returned to baseline within 16-24 h. In vivo imaging of VCAM-1 also demonstrated an acute decrease in expression 4 h after carboplatin administration that recovered within 48 h in mice harboring platinum-resistant tumors. Chronic VCAM-1 expression reflected the effect of platinum-based treatment on tumor burden. Specifically, carboplatin treatment of mice with platinum-sensitive tumors showed reduced VCAM-1 expression, which correlated with reduced tumor burden; mice with platinum-resistant tumors retained elevated VCAM-1 expression and tumor burden after treatment. CONCLUSION: Clinically relevant VCAM-1-specific imaging probes identify VCAM-1 expression as an indicator of ovarian cancer peritoneal metastasis and therapeutic response to platinum-based agents. These observations support testing the utility of VCAM-1 imaging probes to monitor treatment response in ovarian cancer patients, thus providing the potential to improve management of women with this disease.


Subject(s)
Biomarkers, Tumor/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Tomography, Emission-Computed, Single-Photon , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Middle Aged , Neoplasm Metastasis , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/metabolism , Retrospective Studies , Treatment Outcome , Vascular Cell Adhesion Molecule-1/genetics
10.
Gynecol Oncol ; 122(3): 479-83, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21664659

ABSTRACT

OBJECTIVE: Rehospitalization within 30 days of discharge was identified by the Obama Administration as a target for reducing health care spending. We examined readmissions to our gynecologic oncology service to determine: 1) rates of readmission, 2) indication for readmissions, 3) whether the admission was planned, and 4) costs. METHODS: IRB approval was obtained for this 5-year retrospective review (2004-2008). Gynecologic oncology patients were included if they were readmitted within 30 days of discharge at a single academic hospital. Abstracted data included: demographics, dates of hospitalizations, cancer history, indication for admission, and cost. A series of admissions was any number of admissions that occurred within 30 days of discharge. An index admission was the first admission in a series. RESULTS: In the study period, 2455 unique patients were admitted to Gynecologic Oncology. 324 unique patients (13.2%) were readmitted within 30 days, with 37 experiencing >1 series of admission. 87.3% were readmitted to Gynecologic Oncology. Within a series of admissions, patients were admitted on average 1.5 times following the index admission, up to 9 admissions. The median cost of index admission was $9820; for readmissions, $8059. The total cost of readmissions over 5 years was $6,421,733. Unplanned readmissions accounted for the majority of this cost. CONCLUSIONS: Hospital readmissions affect the cost of care, but also the quality of care delivered to our patients. When extrapolated across institutions and across the country, unplanned readmissions are a costly expenditure to patients and the health system, deserving of attention.


Subject(s)
Genital Neoplasms, Female/therapy , Patient Readmission/statistics & numerical data , Female , Genital Neoplasms, Female/economics , Genital Neoplasms, Female/epidemiology , Hospital Costs , Humans , Middle Aged , Patient Readmission/economics , Retrospective Studies , Treatment Outcome , Virginia/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...