Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
3.
Clin Case Rep ; 11(11): e8114, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37908789

ABSTRACT

Key Clinical Message: Isolated utero-ovarian torsion poses a challenge to diagnosing adnexal torsion, as it may not present with imaging findings. Clinicians with high suspicion for torsion but lack of evidence on ultrasonography should proceed to diagnostic laparoscopy. Abstract: Adnexal torsion occurs when the ovary rotates around its supporting ligaments, the infundibulopelvic and utero-ovarian (UO) ligaments, often causing disruption of blood supply. This pathology often presents with acute pelvic pain and is a gynecologic surgical emergency. Diagnosis is typically made with Doppler ultrasound, although dual blood supply to the ovary poses additional diagnostic challenges and sensitivity of this tool is debated. In this case study, we present a case of missed torsion due to isolated compromise of UO ligament.

4.
Mol Cancer Ther ; 22(12): 1404-1412, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37676984

ABSTRACT

Uterine serous carcinoma (USC) is a rare, biologically aggressive variant of endometrial cancer with a high recurrence rate and poor prognosis. HER2 overexpression (3+ positivity) by IHC and/or FISH ERBB2 gene amplification is detected in approximately one-third of patients with USC. Clinical trials incorporating trastuzumab with standard chemotherapy have recently demonstrated improved progression-free and overall survival in advanced-stage or recurrent USC that overexpresses HER2. However, a large number of patients with USC eventually developed resistance to trastuzumab. Trastuzumab deruxtecan (T-DXd) is a novel HER2-directed antibody-drug conjugate with a topoisomerase I inhibitor payload recently approved by the Food and Drug Administration (FDA) for multiple tumor indications. Here, we investigated the in vitro and in vivo efficacy of T-DXd in primary USC cell lines and xenografts with different HER2 expression. T-DXd-induced cell growth suppression in HER2-overexpressing cell lines in vitro, increased early and late apoptosis as assessed by annexin and propidium iodide staining, and, similarly to trastuzumab, T-DXd-induced significant antibody-dependent cellular cytotoxicity in the presence of peripheral blood lymphocytes. While negligible activity was detected against USC cell lines with low HER2 expression, T-DXd demonstrated significant bystander killing against USC tumors with low/negligible HER2 when such cells were admixed with HER2 3+ tumor cells in vitro. T-DXd showed tumor growth suppression in in vivo USC PDX models that overexpress HER2 at 3+ levels, prolonging survival when compared with controls, with minimal toxicity. Future clinical trials are warranted in patients with USC failing trastuzumab treatment.


Subject(s)
Carcinoma , Immunoconjugates , Uterine Neoplasms , Female , Humans , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Camptothecin/pharmacology , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Carcinoma/drug therapy
5.
Cancers (Basel) ; 15(16)2023 Aug 14.
Article in English | MEDLINE | ID: mdl-37627113

ABSTRACT

Uterine serous carcinoma (USC) and uterine carcinosarcoma (UCS) are two rare histologic variants of uterine carcinoma, with distinct molecular profiles and aggressive metastatic potential. As the effectivity of traditional platinum-based chemotherapy for USC and UCS is low, and there are high rates of resistance and recurrence, the development of novel targeted therapeutics is needed. Human epidermal growth factor receptor 2 (HER2) has proven to be an oncogene of increasing interest in these cancers, as HER2 protein overexpression and/or c-ERBB2 gene amplification ranges from ~30 to 35% in USC, and between ~15 and 20% in UCS. This review summarizes the existing clinical and preclinical evidence, as well as ongoing clinical trials of HER2-targeting therapeutics, and identifies potential areas of further development and inquiry.

6.
Ann Surg Oncol ; 30(9): 5597-5609, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37358686

ABSTRACT

Hyperthermic intraperitoneal chemotherapy (HIPEC) is a treatment modality that aims to target the main site of tumor dissemination in ovarian cancer, the peritoneum, by combining the benefits of intraperitoneal chemotherapy with the synergistic effects of hyperthermia all during a single administration at the time of cytoreductive surgery. High-quality evidence currently only supports the use of HIPEC with cisplatin at the time of interval cytoreduction after neoadjuvant chemotherapy for stage III epithelial ovarian cancer. Many questions remain, including HIPEC's role at other timepoints in ovarian cancer treatment, who are optimal candidates, and specifics of HIPEC protocols. This article reviews the history of normothermic and hyperthermic intraperitoneal chemotherapy in ovarian cancer and evidence regarding HIPEC implementation and patient outcomes. Additionally, this review explores details of HIPEC technique and perioperative care, cost considerations, complication and quality of life data, disparities in HIPEC use, and unresolved issues.


Subject(s)
Hyperthermia, Induced , Ovarian Neoplasms , Female , Humans , Hyperthermic Intraperitoneal Chemotherapy , Quality of Life , Hyperthermia, Induced/methods , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/surgery , Carcinoma, Ovarian Epithelial/surgery , Combined Modality Therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytoreduction Surgical Procedures/methods
7.
Int J Mol Sci ; 24(10)2023 May 17.
Article in English | MEDLINE | ID: mdl-37240216

ABSTRACT

Uterine serous carcinoma (USC) and carcinosarcomas (CSs) are rare, highly aggressive variants of endometrial cancer. No reliable tumor biomarkers are currently available to guide response to treatment or detection of early recurrence in USC/CS patients. Circulating tumor DNA (ctDNA) identified using ultrasensitive technology such as droplet digital polymerase chain reaction (ddPCR) may represent a novel platform for the identification of occult disease. We explored the use of personalized ctDNA markers for monitoring USC and CS patients. Tumor and plasma samples from USC/CS patients were collected at the time of surgery and/or during the treatment course for assessment of tumor-specific somatic structural variants (SSVs) by a clinical-grade next-generation sequencing (NGS) platform (i.e., Foundation Medicine) and a droplet digital PCR instrument (Raindance, ddPCR). The level of ctDNA was quantified by droplet digital PCR in plasma samples and correlated to clinical findings, including CA-125 serum and/or computed tomography (CT) scanning results. The genomic-profiling-based assay identified mutated "driver" target genes for ctDNA analysis in all USC/CS patients. In multiple patients, longitudinal ctDNA testing was able to detect the presence of cancer cells before the recurrent tumor was clinically detectable by either CA-125 or CT scanning. Persistent undetectable levels of ctDNA following initial treatment were associated with prolonged progression-free and overall survival. In a USC patient, CA-125 and TP53 mutations but not PIK3CA mutations become undetectable in the plasma at the time of recurrence, suggesting that more than one customized probe should be used for monitoring ctDNA. Longitudinal ctDNA testing using tumor-informed assays may identify the presence of residual tumors, predict responses to treatment, and identify early recurrences in USC/CS patients. Recognition of disease persistence and/or recurrence through ctDNA surveillance may allow earlier treatment of recurrent disease and has the potential to change clinical practice in the management of USC and CS patients. CtDNA validation studies in USC/CS patients prospectively enrolled in treatment trials are warranted.


Subject(s)
Carcinosarcoma , Circulating Tumor DNA , Cystadenocarcinoma, Serous , Uterine Neoplasms , Female , Humans , Circulating Tumor DNA/genetics , Neoplasm Recurrence, Local/genetics , Uterine Neoplasms/diagnosis , Uterine Neoplasms/genetics , Uterine Neoplasms/therapy , Biomarkers, Tumor/genetics , Mutation , Cystadenocarcinoma, Serous/diagnosis , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/therapy , Carcinosarcoma/diagnosis , Carcinosarcoma/genetics , Carcinosarcoma/therapy
8.
Gynecol Oncol ; 172: 65-71, 2023 05.
Article in English | MEDLINE | ID: mdl-36958197

ABSTRACT

INTRODUCTION: Uterine leiomyosarcomas (uLMS) are rare, highly aggressive tumors. Up to 30% of uLMS may harbor gain of function (GOF) in the MAP2K4 gene, important for tumor cell proliferation, differentiation and metastasis. We investigated the in vivo activity of a novel MAP2K4 inhibitor, PLX8725, against uLMS harboring MAP2K4 gene-amplification. METHODS: Two fully characterized uLMS (i.e., LEY-11 and LEY-16) were grafted into female CB-17/SCID mice. Treatments with control vehicle or PLX8725 (50 mg/kg) were given via oral gavage daily on weekdays for up to 60 days. Tumor volume differences were calculated with two-way ANOVA. Pharmacokinetic (PK) and mechanistic studies of PLX8725 in uLMS PDX models were also performed. RESULTS: Both uLMS tumors evaluated demonstrated GOF in MAP2K4 (i.e., 3 CNV in both LEY-11 and LEY-16). Tumor growth inhibition was significantly greater in both PDX LEY-11 and PDX LEY-16 treated with PLX8725 when compared to controls (p < 0.001). Median overall survival was also significantly longer in both PDX LEY-11 (p = 0.0047) and PDX LEY-16 (p = 0.0058) treatment cohorts when compared to controls. PLX8725 oral treatment was well tolerated, and PK studies demonstrated that oral PLX8725 gives extended exposure in mice. Ex vivo tumor samples after PLX8725 exposure decreased phosphorylated-ATR, JNK and p38, and increased expression of apoptotic molecules on western blot. CONCLUSION: PLX8725 demonstrates promising in vivo activity against PDX models of uLMS harboring GOF alterations in the MAP2K4 gene with tolerable toxicity. Phase I trials of PLX8725 in advanced, recurrent, chemotherapy-resistant uLMS patients are warranted.


Subject(s)
Leiomyosarcoma , Pelvic Neoplasms , Uterine Neoplasms , Humans , Female , Animals , Mice , Leiomyosarcoma/drug therapy , Leiomyosarcoma/genetics , Leiomyosarcoma/pathology , Gene Amplification , Mice, SCID , Neoplasm Recurrence, Local/genetics , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , MAP Kinase Kinase 4/genetics
10.
Expert Opin Biol Ther ; 23(3): 227-233, 2023 03.
Article in English | MEDLINE | ID: mdl-36800548

ABSTRACT

INTRODUCTION: Despite progress on prevention and treatment of cervical cancer, global morbidity and mortality remain high. Immunotherapy, in conjunction with standard chemotherapy, presents an opportunity for further benefit. AREAS COVERED: Here we report the pharmacologic properties, evidence for clinical efficacy, safety, and tolerability of pembrolizumab in addition to standard chemotherapy with and without bevacizumab for treatment of advanced or recurrent cervical cancer. EXPERT OPINION: In patients with progressive, recurrent, or metastatic PD-L1 expressing cervical cancer, without contraindication to anti-VEGF therapy, the use of pembrolizumab with bevacizumab and standard chemotherapy with carboplatin and paclitaxel is warranted. There is no evidence to support the use of pembrolizumab for this population broadly, and no evidence to support its use in PD-L1 non-expressing tumors.


Subject(s)
Lung Neoplasms , Uterine Cervical Neoplasms , Female , Humans , Bevacizumab/therapeutic use , Uterine Cervical Neoplasms/drug therapy , B7-H1 Antigen , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Lung Neoplasms/drug therapy
11.
Gynecol Oncol ; 170: 38-45, 2023 03.
Article in English | MEDLINE | ID: mdl-36610380

ABSTRACT

OBJECTIVES: Carcinosarcomas are highly aggressive gynecologic malignancies containing both carcinomatous and sarcomatous elements with heterogeneous HER2/neu expression and limited therapeutic options. We compared the efficacy of trastuzumab deruxtecan (DS-8201a), a novel HER2/neu-targeting antibody-drug conjugate (ADC) to an ADC isotype control (MAAA-9199) against primary uterine and ovarian carcinosarcomas in vitro and in vivo. METHODS: Twelve primary carcinosarcoma (CS) cell lines were evaluated for HER2/neu surface expression by immunohistochemistry (IHC) and by flow cytometry, and gene amplification by fluorescence in situ hybridization (FISH) assays. The in vitro experiments included cytotoxicity and bystander killing effect assays on three cell lines of variable HER2/neu expression. In vivo activity was studied in a mouse CS xenograft model of 3+ HER2/neu uterine CS. RESULTS: In vitro studies showed that DS-8201a was highly effective against uterine and ovarian CS cell lines demonstrating 3+ HER2/neu expression compared to MAAA-9199 control; there was no significant improvement in the 0 HER2/neu CS cell line. However, DS-8201a induced efficient bystander killing of 0 HER2/neu tumor cells when admixed with 3+ HER2/neu cells. In vivo studies confirmed that DS-8201a was more effective than MAAA-9199 in 3+ HER2/neu-expressing CS xenografts. CONCLUSION: DS-8201a may represent a novel and highly effective ADC against HER2/neu-expressing CS.


Subject(s)
Carcinosarcoma , Immunoconjugates , Ovarian Neoplasms , Humans , Female , Mice , Animals , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/therapeutic use , In Situ Hybridization, Fluorescence , Receptor, ErbB-2/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Line, Tumor , Trastuzumab/therapeutic use , Immunoconjugates/therapeutic use , Ovarian Neoplasms/pathology , Carcinosarcoma/pathology
12.
Gynecol Oncol ; 170: 172-178, 2023 03.
Article in English | MEDLINE | ID: mdl-36706643

ABSTRACT

INTRODUCTION: Ovarian cancer (OC) is associated with the highest gynecologic cancer mortality. The development of novel, effective combinations of targeted therapeutics remains an unmet medical need. We evaluated the preclinical efficacy of the Poly (ADP-ribose) polymerase (PARP) inhibitor (olaparib) and the pan-ErbB inhibitor (neratinib) as single agents and in combination in ovarian cancer cell lines and xenografts with variable HER2 expression. METHODS: In vitro cell viability with olaparib, neratinib, and their combination was assessed using flow-cytometry based assays against a panel of OC primary cell lines with variable HER2 expression. Immunoblotting experiments were performed to elucidate the mechanism of activity and synergism. The in vivo antitumor activity of the olaparib/neratinib combination versus single agents was tested in HER2 positive xenograft OC models. RESULTS: HER2 + OC cell lines demonstrated higher sensitivity to olaparib and neratinib when compared to HER2 negative tumors (i.e., IC50: 2.06 ± 0.33 µM vs. 39.28 ± 30.51 µM, p = 0.0035 for olaparib and 19.42 ± 2.63 nM vs. 235.0 ± 165.0 nM, p = 0.0035 for neratinib). The combination of olaparib with neratinib was more potent when compared to single-agent olaparib or neratinib both in vitro and in vivo, and demonstrated synergy in all primary HER2 + OC models. Western blot experiments showed neratinib decreased pHER2/neu while increased Poly(ADP-ribose) (PAR) enzymatic activity; olaparib increased pHER2/Neu expression and blocked PAR activatio. Olaparib/neratinib in combination decreased both pHER2/Neu as well as PAR activation. CONCLUSION: The combination of olaparib and neratinib is synergistic and endowed with remarkable preclinical activity against HER2+ ovarian cancers. This combination may represent a novel therapeutic option for ovarian cancer patients with HER2+, homologous recombination-proficient tumors resistant to chemotherapy.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Humans , Female , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Ribose/therapeutic use , Antineoplastic Agents/therapeutic use , Phthalazines/therapeutic use , Ovarian Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/therapeutic use , Cell Line, Tumor
13.
Gynecol Oncol ; 169: 98-105, 2023 02.
Article in English | MEDLINE | ID: mdl-36525930

ABSTRACT

BACKGROUND: Carcinosarcoma of the ovary (OCS) and uterus (UCS) are rare highly aggressive malignancies. Ataxia-telangiectasia-and-Rad3-related (ATR) kinase and homologous recombination play a pivotal role in DNA damage repair. Homologous recombination deficiency (HRD) has been demonstrated in >30% of OCS/UCS. We investigated the preclinical activity of elimusertib, a selective ATR kinase inhibitor, against carcinosarcoma (CS) cell lines and xenografts. METHODS: Sensitivity to elimusertib was evaluated in vitro against nine whole exome-sequenced (WES) primary CS cell lines and in vivo against HRD CS xenografts. Western blots were performed to determine baseline ATR and p-ATR protein expression in CS, and ATR pathway downstream effectors and apoptosis markers in CS HRD cell lines after Elimusertib treatment. RESULTS: Out of the 9 CS cell lines, 3 harbored HRD and 6 homologous recombination proficient (HRP) features. Most of CS (i.e., 7/9 = 85%) were found to be sensitive to Elimusertib in vitro. Among the 5 primary CS cell lines with a high-grade pure serous epithelial component, HRD cell lines were more sensitive to elimusertib than HRP tumors (mean IC50 ± SEM HRD CS = 61.3 nM ±15.2 vs HRP = 361.6 nM ±24.4 (p = 0.01)). Baseline ATR and p-ATR protein expression was higher in HRD CS cell lines. Elimusertib showed tumor growth inhibition in HRD CS xenografts (p < 0.0001) and increased overall animal survival (p < 0.0001). Western blot demonstrated dose-dependent inhibition of ATR, p-ATR and its downstream effector p-CHK1, and a dose-dependent increase in caspase-3 expression. CONCLUSIONS: Elimusertib is preclinically active in vitro and in vivo against primary CS cell lines and xenografts, respectively. CS models harboring HRD or with pure/mixed endometrioid histology demonstrated higher sensitivity to ATR inhibition. Clinical trials with elimusertib in CS patients are warranted.


Subject(s)
Antineoplastic Agents , Ataxia Telangiectasia , Carcinosarcoma , Uterine Neoplasms , Female , Animals , Humans , Ataxia Telangiectasia/drug therapy , Ovary , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line, Tumor , Antineoplastic Agents/therapeutic use , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Carcinosarcoma/drug therapy , Carcinosarcoma/genetics
14.
Gynecol Oncol ; 168: 157-165, 2023 01.
Article in English | MEDLINE | ID: mdl-36442427

ABSTRACT

INTRODUCTION: Uterine leiomyosarcoma (uLMS) is a rare, highly aggressive malignancy. Recent data suggest 50% of uLMS may harbor alterations in the ATRX gene and such mutations may confer sensitivity to ataxia-telangiectasia-and-Rad3-related (ATR) kinase inhibitors. We sought to investigate the in vivo activity of Elimusertib (BAY1895344), a novel ATR-inhibitor, against ATRX-mutated uLMS patient-derived xenografts (PDXs). METHODS: Two fully characterized uLMS (i.e., LEY-11 and LEY-16) were grafted into female CB-17/SCID mice. Treatments with control vehicle or BAY1895344 (20 mg/kg dosed twice daily 3 days on 4 days off) were given via oral gavage and tumor measurements as well as weights obtained twice weekly. Tumor volume differences were calculated with a two-way ANOVA. Mechanistic studies were performed ex vivo using BAY1895344 treated uLMS tumor samples by western blot analysis. RESULTS: Both PDX LEY-11 and PDX LEY-16 harboring ATRX gene mutations demonstrated an aggressive behavior in vivo (i.e., control mice were euthanized on average at day 12.5 for PDX LEY-11 and at day 33 for PDX LEY-16). In both tumor models BAY1895344 20 mg/kg dosed with an intermittent oral schedule was able to induce significant growth inhibition compared to vehicle control treatment (p < 0.001 for both LEY-11 and LEY-16) and prolong median overall survival [PDX LEY-11 (12.5 vs. 42 days, p < 0.001) and PDX LEY-16 (33 vs. 60 days, p < 0.001)]. There were not significant changes in weight between treatment and controls. By western blot assays BAY1895344 exposure decreased phosphorylated-ATR and increased expression of apoptotic molecules in LMS PDXs. CONCLUSIONS: BAY1895344 demonstrates promising in vivo activity against biologically aggressive PDX models of uLMS harboring ATRX mutations, with no significant toxicity. Clinical trials of BAY1895344 in uLMS patients are warranted.


Subject(s)
Leiomyosarcoma , Uterine Neoplasms , Humans , Female , Animals , Mice , Leiomyosarcoma/drug therapy , Leiomyosarcoma/genetics , Leiomyosarcoma/pathology , Mice, SCID , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Mutation , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics
15.
Gynecol Oncol ; 166(2): 351-357, 2022 08.
Article in English | MEDLINE | ID: mdl-35641325

ABSTRACT

INTRODUCTION: Uterine serous carcinoma (USC) is an aggressive variant of endometrial cancer with a poor prognosis. Approximately 30% of USC overexpress HER2/neu, a recognized target for trastuzumab in advanced/recurrent HER2/neu-positive USC. We evaluated the efficacy of the pan-c-erb inhibitor neratinib and the poly (ADP-ribose)-polymerase (PARP) inhibitor olaparib as single agents and in combination against USC cell lines and xenografts. METHODS: In-vitro cell-viability assays with olaparib, neratinib, and olaparib/neratinib were assessed using flow-cytometry based assays against a panel of USC cell lines with high and low HER2/neu expression. Homologous recombination deficiency (HRD) signatures were evaluated as described by Alexandrov et al. (Nature;2020;578:94-101) while downstream signaling affected by neratinib/olaparib exposure was assessed with immunoblotting. Efficacy of single- versus dual-agent inhibition was evaluated in-vivo using two USC-xenografts with 3+ HER2/neu expression. RESULTS: Neratinib was more potent than olaparib in suppression of in-vitro growth of HER2/neu 3+ cell lines (ARK1: p = 0.0047; ARK2: p = 0.0428) while no difference was noted against HER2/neu 1+ tumors (ARK4). Importantly, the combination of olaparib with neratinib synergistically improved tumor suppression compared to either single-agent in vitro. USC cells exposed to olaparib upregulated HER2/neu expression, while neratinib treatment increased PARP activity (ARK1: p < 0.0001; ARK2: p < 0.0001). Single-agent neratinib transiently inhibited in vivo growth of USC xenografts harboring HER2/neu gene amplification (ARK1: p < 0.05; ARK2: p < 0.05). In contrast, the combination of the two inhibitors caused a stronger and durable growth inhibition in both USC xenografts (ARK1: p < 0.05; ARK2: p < 0.05). CONCLUSION: The combination of olaparib and neratinib is active and synergistic against primary HER2/neu + USC. This combination may represent a novel therapeutic option for USC patients with HER2/neu+, homologous recombination-proficient tumors resistant to chemotherapy.


Subject(s)
Cystadenocarcinoma, Serous , Uterine Neoplasms , Cell Line, Tumor , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Female , Humans , Phthalazines , Piperazines , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Quinolines , Receptor, ErbB-2/metabolism , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Xenograft Model Antitumor Assays
16.
Expert Rev Anticancer Ther ; 22(6): 633-645, 2022 06.
Article in English | MEDLINE | ID: mdl-35533682

ABSTRACT

INTRODUCTION: Cervical cancer is the overall fourth most common malignancy and the fourth most common cause of cancer-related deaths worldwide. Despite vaccination and screening programs, many women continue to present with advanced stage cervical cancer, wherein the treatment options have been limited. AREAS COVERED: In this review, immunotherapy and the potential targeted therapies that have demonstrated promise in the treatment of persistent, recurrent, and metastatic cervical cancer are discussed. EXPERT OPINION: Our global goal in the gynecologic oncology community is to eliminate cervical cancer, by increasing the uptake of preventive vaccination and screening programs. For unfortunate patients who present with metastatic, persistent, and recurrent cervical cancer, pembrolizumab with chemotherapy, with or without bevacizumab is the new first-line therapy for PD-L1 positive patients. For this patient population as a second-line therapy, tisotumab vedotin (i.e. ADC) has shown significant efficacy in phase II trials, leading to the US Food and Drug Administration approval. Combination regimens inclusive of immune checkpoint inhibitors, DNA damage repair inhibitors, and antibody drug conjugates are potential breakthrough treatment strategies and are currently being investigated.


Subject(s)
Uterine Cervical Neoplasms , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Female , Humans , Immunotherapy , Neoplasm Recurrence, Local , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology
17.
Gynecol Oncol ; 166(1): 117-125, 2022 07.
Article in English | MEDLINE | ID: mdl-35599167

ABSTRACT

OBJECTIVES: Carcinosarcoma (CS) of the ovary and uterus are highly aggressive malignancies associated with poor survival. Poly(ADP-ribose)-polymerase inhibitors (PARPi) are targeted agents impairing DNA repair via homologous-recombination-deficiency (HRD) mechanisms. We used whole-exome-sequencing (WES) data from a cohort of fresh tumor samples of ovarian (OCS) and uterine carcinosarcoma (UCS), primary cell lines and xenografts to investigate the role for olaparib in CSs. METHODS: WES data from 73 CS samples (48 UCS and 25 OCS) were analyzed for HRD signatures. Olaparib activity was evaluated using cell-viability, cell-cycle, apoptosis and cytotoxicity assays against primary CS cell lines. Olaparib antitumor activity was tested in vivo against HRD CS xenografts. RESULTS: Signature-3 (i.e. HRD-related signature) was identified in 60% of OCS (15 of 25) vs 25% of UCS (12 of 48) (p = 0.005). CS cell lines harboring Signature-3/HRD (3 OCS/1 UCS) were significantly more sensitive to olaparib when compared to HRP cell lines (5 UCS/1 OCS) [mean IC50 ± SEM = 2.94 µM ± 0.07 vs mean ± SEM = 23.3 µM ± 0.09, (p = 0.02), respectively]. PARPi suppressed CS cell growth through cell cycle arrest in the G2/M phase and caused more apoptosis in HRD vs HRP primary tumors (p < 0.0001). In vivo, olaparib significantly impaired HRD CS xenografts tumor growth (p = 0.0008) and increased overall animal survival (p < 0.0001). CONCLUSIONS: OCS and UCS cell lines harboring HRD signature-3 were significantly more sensitive to olaparib in vitro and in vivo when compared to HRP CS. Clinical studies with PARPi in CS patients with a dominant signature 3 (HRD-related) are warranted.


Subject(s)
Carcinosarcoma , Ovarian Neoplasms , Adenosine Diphosphate/therapeutic use , Animals , Carcinosarcoma/drug therapy , Carcinosarcoma/genetics , Cell Line, Tumor , Female , Homologous Recombination , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovary/pathology , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerases , Ribose/therapeutic use
18.
BMJ Case Rep ; 15(4)2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35383096

ABSTRACT

A 71-year-old woman presented with an intravesical bladder mass found to be a clear cell adenocarcinoma of Müllerian origin with positive PAX-8 staining after transurethral resection. Partial cystectomy along with total hysterectomy were performed, and final pathology revealed no residual tumour and extensive endometriosis. She declined adjuvant therapy and was dispositioned to surveillance.


Subject(s)
Adenocarcinoma, Clear Cell , Urinary Bladder Diseases , Urinary Bladder Neoplasms , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/surgery , Aged , Cystectomy , Female , Humans , Urinary Bladder/pathology , Urinary Bladder/surgery , Urinary Bladder Diseases/surgery , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery
19.
Gynecol Oncol Rep ; 40: 100959, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35372655

ABSTRACT

•Extensive ureterolysis due to the lateral extent of the tumor with postoperative ureteral stenting.•Partial urinary bladder cystectomy due to clinical concern for invasion followed by two-layer cystorrhapy.•'Port-hopping' for development of the bilateral tunnels of Wertheim with the Vessel Sealer Extend.

20.
Expert Rev Anticancer Ther ; 22(3): 249-258, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35176955

ABSTRACT

INTRODUCTION: Endometrial cancer (EC) is the most common gynecologic malignancy. Outcomes for patients with advanced and/or recurrent disease have been modest with the use of chemotherapy. The approval of immune checkpoint inhibitors targeting PD-1 has recently revolutionized human cancer treatment. Recent trials with immune checkpoint inhibitors used alone or in combination with other agents, have demonstrated remarkable efficacy in the treatment of the all-comers EC patient population. AREAS COVERED: In this article, we review major clinical trials on PD-1/PD-L1 inhibitors in advanced and recurrent EC and discuss the response rates of these agents in the context of their genomic background. EXPERT OPINION: Immune checkpoint inhibitors have significantly changed our approach to the treatment of advanced/recurrent EC. Single agent anti-PD-1 regimens are highly effective in MMRd/MSI-H patients, but their clinical efficacy remains modest in MMR proficient/TMB low EC patients. Combination regimens that can decrease the tumor microenvironments immunosuppression and increase tumor immunogenicity represent a viable treatment option to broaden the activity of immune checkpoint inhibitors in advanced/recurrent EC patients. An increased understanding of the biomarkers of response and the molecular mechanisms of resistance to immune checkpoint inhibitors remains key for the next advancement of the field.


Subject(s)
Endometrial Neoplasms , Immune Checkpoint Inhibitors , Endometrial Neoplasms/drug therapy , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Microsatellite Instability , Neoplasm Recurrence, Local/drug therapy , Programmed Cell Death 1 Receptor , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...