Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Clin Cancer Res ; 28(5): 948-959, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34907079

ABSTRACT

PURPOSE: Advances in our understanding of the contribution of aberrant glycosylation to the pro-oncogenic signaling and metastasis of tumor cells have reinvigorated the development of mucin-targeted therapies. Here, we validate the tumor-targeting ability of a novel monoclonal antibody (mAb), AR9.6, that binds MUC16 and abrogates downstream oncogenic signaling to confer a therapeutic response. EXPERIMENTAL DESIGN: The in vitro and ex vivo validation of the binding of AR9.6 to MUC16 was achieved via flow cytometry, radioligand binding assay (RBA), and immunohistochemistry (IHC). The in vivo MUC16 targeting of AR9.6 was validated by creating a 89Zr-labeled radioimmunoconjugate of the mAb and utilizing immunoPET and ex vivo biodistribution studies in xenograft models of human ovarian and pancreatic cancer. RESULTS: Flow cytometry, RBA, and IHC revealed that AR9.6 binds to ovarian and pancreatic cancer cells in an MUC16-dependent manner. The in vivo radiopharmacologic profile of 89Zr-labeled AR9.6 in mice bearing ovarian and pancreatic cancer xenografts confirmed the MUC16-dependent tumor targeting by the radioimmunoconjugate. Radioactivity uptake was also observed in the distant lymph nodes (LNs) of mice bearing xenografts with high levels of MUC16 expression (i.e., OVCAR3 and Capan-2). IHC analyses of these PET-positive LNs highlighted the presence of shed antigen as well as necrotic, phagocytized, and actively infiltrating neoplastic cells. The humanization of AR9.6 did not compromise its ability to target MUC16-expressing tumors. CONCLUSIONS: The unique therapeutic mechanism of AR9.6 combined with its excellent in vivo tumor targeting makes it a highly promising theranostic agent. huAR9.6 is poised for clinical translation to impact the management of metastatic ovarian and pancreatic cancers.


Subject(s)
Immunoconjugates , Ovarian Neoplasms , Pancreatic Neoplasms , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , CA-125 Antigen , Carcinogenesis , Cell Line, Tumor , Female , Humans , Immunoconjugates/therapeutic use , Membrane Proteins/metabolism , Mice , Mucins/metabolism , Ovarian Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Radioisotopes/therapeutic use , Tissue Distribution , Zirconium , Pancreatic Neoplasms
3.
Gynecol Oncol ; 156(3): 523-529, 2020 03.
Article in English | MEDLINE | ID: mdl-31916979

ABSTRACT

BACKGROUND: This randomized phase II study tested the hypothesis that schedule dependent chemo-immunotherapy with oregovomab improves progression free survival (PFS) and overall survival (OS) in optimally resected, Stage III/IV ovarian cancer. METHODS: Patients from both academic centers and private practice in the US and Italy with Stage III/IV optimally cytoreduced ovarian cancer were randomized to standard six cycle IV carboplatin-paclitaxel chemotherapy (CP) versus CP plus four immunizations with oregovomab (CPO). A translational assessment of a cellular immune response was the primary endpoint; PFS and OS were measured as secondary endpoints. FINDINGS: 97 patients at thirteen centers were accrued to the protocol, 47 to CPO and 50 to CP. Technical issues led to inconsistent performance of the primary CA125 ELISPOT leading to unevaluable results. At a median follow up of 42 months, PFS and OS outcomes revealed an unexpectedly large treatment effect for CPO relative to CP alone, with median PFS of 41.8 months (95% C.I.: 21.8 - N.E.) for CPO and 12.2 months (10.4-18.6) for CP (p = 0.0027, HR 0.46, CI 0.28-0.7). For OS, the median for CPO has not yet been reached (NE) (45.2-NE) and for CP was 43.2 months (31.8-NE) (p = 0.043, HR 0.35, CI 0.16-0.74). The oregovomab treatment resulted in no change in toxicity profile from CP. INTERPRETATION: The previously identified potential clinical benefit of IV CP when administered with oregovomab was further refined in this randomized phase II study. Increases of PFS and OS of statistically and clinically significant magnitude were evident in this study of a front line chemo-immunotherapy treatment of ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Carboplatin/administration & dosage , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/pathology , Combined Modality Therapy , Female , Humans , Immunotherapy/methods , Middle Aged , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Treatment Outcome
4.
Cancer Immunol Immunother ; 69(3): 383-397, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31897661

ABSTRACT

The standard-of-care (SOC) first-line therapy for ovarian cancer (OC) patients is plagued with high relapse rates. Several studies indicated the immune system's prominent role changing the disease course in OC patients. Chemo-immunotherapy regimens, currently being explored, include oregovomab, which is a monoclonal antibody specific for the OC associated antigen carbohydrate/cancer antigen 125 (CA125) that yielded promising results when administered together with SOC in a previous study. The QPT-ORE-002 multi-site phase II randomized study demonstrated that in patients with advanced OC, oregovomab combined with first-line SOC improved overall and progression-free survival, compared to SOC alone. The study included an Italian cohort in which we demonstrated that adding oregovomab to SOC resulted in increased patient numbers with amplified CA125-specific CD8+T lymphocytes/ml peripheral blood counts, which might explain the improved therapeutic effect of SOC + oregovomab over SOC alone. Predictive for oregovomab efficacy was a less suppressive immune environment at baseline as indicated by low numbers of circulating myeloid-derived suppressor cells, subset type 4, and a low neutrophil-and-monocyte to lymphocyte ratio.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/therapeutic use , Immunotherapy/methods , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carboplatin/pharmacology , Female , Humans , Middle Aged , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Precision Medicine
5.
Am J Clin Oncol ; 42(10): 755-760, 2019 10.
Article in English | MEDLINE | ID: mdl-31513018

ABSTRACT

OBJECTIVE: Cancer antigen (CA)-125 influences progression, metastasis, and outcomes in pancreatic cancer. This phase I/II trial (NCT01959672) evaluated the safety, efficacy, and immunologic correlates of chemoimmunotherapy (CIT) with oregovomab (anti-CA-125), followed by stereotactic body radiotherapy (SBRT) with the radiosensitizer nelfinavir. MATERIALS AND METHODS: Following imaging, pathologic confirmation, and staging laparoscopy, subjects received three 3-week cycles of CIT (gemcitabine/leucovorin/fluorouracil/oregovomab). Thereafter, nelfinavir was delivered (1250 mg bid) for 5 weeks, with SBRT (40 Gy/5 fractions) occurring during the third week of nelfinavir. Following another cycle of CIT, pancreaticoduodenectomy was performed if resectable. Three more cycles of CIT were then delivered (total 7 cycles). In subjects with high (≥10 U/mL) CA-125, oregovomab (2 mg) was administered for 7 total doses (3 pre-SBRT, 1 between SBRT and resection, and 3 postoperatively). The enzyme-linked immunospot assay evaluated the development of CA-125-specific CD8 T-lymphocytes. RESULTS: The trial was prematurely closed because gemcitabine/leucovorin/fluorouracil was replaced by FOLFIRINOX and gemcitabine/nab-paclitaxel as the standard of care. Median follow-up was 13 months. Of 11 enrolled patients, 10 had high CA-125; 1 patient suffered an unexpected cardiac-related death, so 9 subjects received oregovomab. Ten received SBRT and 4 underwent resection. Overall, 6/11 patients experienced any grade ≥3 event. The median survival and time to progression were 13 and 8.6 months, respectively. Five patients had samples available for immunospot testing, of whom 2 (40%) developed CA-125-specific CD8 T-lymphocytes. CONCLUSION: A combined pancreatic cancer multimodality approach using CIT and radiosensitized radiotherapy is feasible and safe; delivery of immunotherapy can lead to T-cell immunity. Re-evaluation with modern systemic paradigms is recommended.


Subject(s)
Adenocarcinoma/therapy , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Nelfinavir/therapeutic use , Neoadjuvant Therapy/methods , Pancreatic Neoplasms/therapy , Radiosurgery/methods , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CA-125 Antigen/blood , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Combined Modality Therapy , Disease-Free Survival , Female , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Leucovorin/adverse effects , Leucovorin/therapeutic use , Magnetic Resonance Imaging/methods , Male , Membrane Proteins/blood , Middle Aged , Nelfinavir/adverse effects , Neoplasm Invasiveness/pathology , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/therapeutic use , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreaticoduodenectomy/methods , Proportional Hazards Models , Risk Assessment , Survival Analysis , Time Factors , Tomography, X-Ray Computed/methods , Treatment Outcome
6.
Mater Sci Eng C Mater Biol Appl ; 77: 935-946, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28532114

ABSTRACT

A nanoparticulate photodynamic approach was employed with an objective to achieve enhanced production of singlet oxygen (1O2), for the management of posterior segment eye diseases like age related macular degeneration. The hypocrellin B (HB) loaded poly lactide-co-glycolide nanoparticle formulations were incorporated with nano silver (HBS-NPs). The optimized HBS-NPs contained 2.60±0.06mg/mL of HB and showed (i) 135.6 to 828.2nm size range, and (ii) negative zeta potential with a narrow polydispersity index. The DSC thermograms suggested the amorphous nature of HB inside the HBS-NPs. With the average encapsulation efficiency of 92.9±1.79%, the drug release from the HBS-NPs followed a biphasic pattern with an initial burst of 3.50% during first 8h followed by a sustained release of 47.82% within 3days. The interaction between nano silver and HB as assessed by the increase in spectral intensity of Raman spectrum demonstrates that HB may be attached over the nano silver. Generation of reactive oxygen species (ROS) by HBS-NPs was significantly higher than that of HB/HB-NPs. The singlet oxygen generating efficiency assessed using EPR spectrometer follows the order of nano silver>HB-NPs>pure HB drug solution>HBS-NPs. The HBS-NPs had a concentration and time dependent phototoxicity on A549 (human adeno lung carcinoma) cells in the presence of light providing a superior phototoxic effect (82.2% at 50µM) at 2h irradiation. The CAM treated with HBS-NPs showed a significant anti-angiogenic effect compared to a blank formulation. In vivo biodistribution studies revealed that intravenous administration of HBS-NPs lead into significant exposure to the posterior segment of the eye. This proof of principle study demonstrates that HB based nanoparticles may be a valuable new tool for application in ocular photodynamic therapy for the treatment of AMD in future.


Subject(s)
Metal Nanoparticles , A549 Cells , Humans , Perylene/analogs & derivatives , Photochemotherapy , Quinones , Silver , Singlet Oxygen , Tissue Distribution
7.
Invest New Drugs ; 29(6): 1328-36, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20676746

ABSTRACT

Development of sonosensitizers for sonodynamic therapy (SDT) which selectively target abnormal cells can limit undesired side effects in chemotherapeutic applications. Hypocrellin-B (HB) derivatives are low molecular weight compounds which belong to the perylenequinone family of photosensitizing and sonosensitizing compounds. In this study, we investigate the cytotoxic mechanisms of a novel HB-derived photo- and sonosensitizer, SL017. Human fibroblast WI-38 cells were treated with SL017 (0 µM, 0.1 µM or 10 µM) and subjected to photodynamic therapy (PDT) or SDT. Studies demonstrate that maximal uptake of SL017 occurs within 30 min, with a mitochondrial subcellular localization. Activation of SL017 by either visible light or ultrasound resulted in significant increases in reactive oxygen species (ROS) production as measured by CM-H2-DCFDA (5-(and-6)-chloromethyl-2'7'-dichlorodihydrofluorescein diacetate acetyl ester). Co-administration of the antioxidant, ascorbic acid, attenuated ROS production. Low concentrations of SL017 (100 nM) induced a rapid (<90 s) loss of mitochondrial membrane potential (ΔΨm). Epoxyeicosatrienoic acids (EETs), cytochrome P450-derived metabolites of arachidonic acid (AA) involved in maintaining homeostasis and protection against cell injury, were able to attenuate loss of ΔΨm, however ascorbic acid was not. SL017 treatment resulted in increased mitochondrial fragmentation which followed loss of ΔΨm. Our studies demonstrate that SL017 targets mitochondria, triggering collapse of mitochondrial membrane potential, generates ROS and subsequently results in mitochondrial fragmentation.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Perylene/analogs & derivatives , Photosensitizing Agents/pharmacology , Quinones/pharmacology , 8,11,14-Eicosatrienoic Acid/pharmacology , Cell Death/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Perylene/administration & dosage , Perylene/pharmacology , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Quinones/administration & dosage , Reactive Oxygen Species/metabolism , Ultrasonic Therapy/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...