Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Anticancer Res ; 44(7): 2765-2768, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925853

ABSTRACT

A "Think Tank for Osteosarcoma" medical advisory board meeting was held in Santa Monica, CA, USA on February 2-3, 2024. The goal was to develop a strategic approach to prevent recurrence of osteosarcoma. Osteosarcoma metabolism and the genomic instability of osteosarcoma, immunotherapy for osteosarcoma, CAR-T cell therapy, DeltaRex-G tumor-targeted gene therapy, repurposed drugs, alternative medicines, and personalized medicine were discussed. Only DeltaRex-G was voted on. The conclusions were the following: No intervention has been demonstrated to improve survival in a clinical trial. Additionally, the consensus (10/12 in favor) was that DeltaRex-G without immunotherapy may be administered for up to one year. Phase 2/3 randomized studies of DeltaRex-G should be performed to determine whether the incidence of recurrence could be reduced in high-risk individuals. Furthermore, a personalized approach using drugs with minimal toxicity could be attempted with the acknowledgement that there are no efficacy data to base this on. Repurposed drugs and alternative therapies should be tested in mouse models of osteosarcoma. Moreover, unmodified IL-2 primed Gamma Delta (NK) cell therapy may be used to prevent recurrence. Lastly, rapid development of CAR-T cell therapy is recommended, and an institute dedicated to the study of osteosarcoma is needed.


Subject(s)
Bone Neoplasms , Osteosarcoma , Osteosarcoma/therapy , Osteosarcoma/pathology , Humans , Bone Neoplasms/therapy , Bone Neoplasms/pathology , Animals , Immunotherapy/methods , Precision Medicine/methods , Advisory Committees , Neoplasm Recurrence, Local/prevention & control , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy
2.
J Clin Oncol ; 42(13): 1472-1476, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38427923

ABSTRACT

Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported.nab-Sirolimus is approved in the United States for the treatment of metastatic or locally advanced malignant perivascular epithelioid cell tumor (PEComa) on the basis of the primary analysis results of the phase II Advanced Malignant Perivascular Epithelioid Cell Tumors (AMPECT) trial (ClinicalTrials.gov identifier: NCT02494570). Results from the primary analysis were previously published; however, the median duration of response (mDOR) had not been reached at that time. Here, 3 years after the primary analysis, we report final efficacy and safety data (data cutoff: April 29, 2022). At study completion, the confirmed overall response rate (by independent radiologist review using RECIST v1.1) was 38.7% (95% CI, 21.8 to 57.8), with an additional converted confirmed complete response (n = 2). Median progression-free survival remained the same at 10.6 months (95% CI, 5.5 to 41.2). The mDOR was reached at 39.7 months (95% CI, 6.5 to not reached [NR]), and the median overall survival at completion was 53.1 months (95% CI, 22.2 to NR). The most common treatment-related adverse events (TRAEs) were stomatitis (82.4%) and fatigue and rash (each 61.8%). No new or unexpected adverse events occurred, and no grade ≥4 TRAEs were reported. These results highlight the long-term clinical benefit of nab-sirolimus in patients with advanced malignant PEComa, with a DOR of >3 years.


Subject(s)
Perivascular Epithelioid Cell Neoplasms , Sirolimus , Humans , Female , Male , Middle Aged , Perivascular Epithelioid Cell Neoplasms/drug therapy , Adult , Aged , Sirolimus/therapeutic use , Sirolimus/adverse effects , Sirolimus/administration & dosage , Progression-Free Survival , Antibiotics, Antineoplastic/therapeutic use , Antibiotics, Antineoplastic/adverse effects
4.
Front Oncol ; 13: 1116937, 2023.
Article in English | MEDLINE | ID: mdl-37234994

ABSTRACT

Background: Intratumoral injection of talimogene laherparepvec evokes a cytotoxic immune response. Therefore, the combination of talimogene laherparepvec with trabectedin and nivolumab may have synergistic effects in advanced sarcomas. Patients and methods: This phase 2 trial was conducted from May 30, 2019 to January 31, 2022. Endpoints: Primary: Progression free survival rate at month 12. Secondary: Best overall response, progression free survival rate at 6 and 9 months, overall survival rate at 6, 9, and 12 months, incidence of conversion of an unresectable tumor to a resectable tumor, and incidence of adverse events. Eligible patients had to be ≥ 18 years of age, have advanced histologically proven sarcoma, at least 1 previous chemotherapy regimen, and at least one accessible tumor for intratumoral injection. Treatment: Trabectedin intravenously (1.2 mg/m2 q3 weeks), nivolumab intravenously (3 mg/kg q2 weeks), and intratumoral talimogene laherparepvec (1x108 plaque forming units/ml q2 weeks). Results: Median time of follow-up: 15.2 months. Efficacy analysis: Thirty-nine patients who had completed at least one treatment cycle and had a follow-up computerized tomography were evaluable for efficacy analysis. Median number of prior therapies: 4 (range 1-11). Progression free survival rate at month 12, 36.7%. Confirmed Best Overall Response by Response Evaluation Criteria in Solid Tumors v1.1 = 3 partial responses, 30 stable disease, 6 progressive disease. Best Overall Response Rate, 7.7%, Disease Control Rate, 84.6%; median progression free survival, 7.8 (95% Confidence Intervals: 4.1-13.1) months; 6-, 9-, 12-month progression free survival rates, 54.5%/45.9%/36.7%; median overall survival 19.3 (95% Confidence Intervals: 12.8 -.) months; 6-, 9- and 12-month overall survival rate, 86.9%/73.3%/73.3%. One patient had a complete surgical resection. Fifty percent of patients had a ≥ grade 3 treatment related adverse events which included anemia (6%), thrombocytopenia (6%), neutropenia (4%), increased alanine transaminase (4%), decreased left ventricular ejection fraction (4%), dehydration (4%), hyponatremia (4%). Conclusions: Taken together these data suggest that the TNT regimen is effective and safe for advanced previously treated sarcomas, and is worth being further studied in a randomized phase 3 trial as first- or second- line treatment for patients with advanced sarcomas.

5.
Anticancer Res ; 43(6): 2383-2391, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37247916

ABSTRACT

Women with HR+HER2+ early-stage breast cancer are disadvantaged by the lack of clinical trials focused on women ≥70 years of age. In the past years, there has been increasing controversy on the use of toxic chemotherapy as standard of care treatment for early- stage HR+ HER2+ breast carcinoma in older women. With precision medicine coming of age, molecular profiling of tumors and circulating tumor DNA has identified target oncogenes that could be used in designing an optimal treatment for this group of women. This article reviews the current treatment of early-stage triple receptor positive breast cancer, the risks of chemotherapy in older women, and CCNG1, a novel biomarker in development for the use of DeltaRex-G, a CCNG1 inhibitor. Further, future perspectives for DeltaRex-G in older women with early stage CCNG1+ HR+ HER2+ breast cancer are discussed.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2 , Female , Humans , Aged , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Trastuzumab , Cyclin G1
6.
Anticancer Res ; 43(5): 1993-2002, 2023 May.
Article in English | MEDLINE | ID: mdl-37097693

ABSTRACT

BACKGROUND/AIM: Advanced sarcoma has a poor prognosis. Dysregulation of the mammalian target of rapamycin (mTOR) occurs in various types of cancer. We aimed to determine the safety and efficacy of mTOR inhibitor nab-sirolimus when combined with the immune checkpoint inhibitor nivolumab. PATIENTS AND METHODS: Previously treated patients ≥18 years with confirmed diagnosis of advanced sarcoma or tumor with mutations in the mTOR pathway were treated with 3 mg/kg nivolumab intravenously every 3 weeks; escalating doses of nab-sirolimus at 56, 75 or 100 mg/m2 were administered intravenously on days 8 and 15 beginning in cycle 2. The primary aim was to determine the maximum-tolerated dose; we also determined disease control, objective response, progression-free survival, overall survival, and correlation between response using Immune-related Response Evaluation Criteria for Solid Tumors (irRECIST) versus RECIST v1.1. RESULTS: The maximum-tolerated dose was 100 mg/m2 There were two patients with partial response, 12 with stable disease and 11 with progressive disease. Median progression-free and overall survival were 12 and 47 weeks, respectively. The best responders (partial responses) were patients with undifferentiated pleomorphic sarcoma with loss of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), tuberous sclerosis complex 2 (TSC2) mutation and estrogen receptor-positive leiomyosarcoma. Treatment-related adverse events of grade 3 or more included thrombocytopenia, oral mucositis, rash, hyperlipidemia and increased serum alanine aminotransferase. CONCLUSION: The data indicate that (i) treatment with nivolumab plus nab-sirolimus is safe with no unexpected adverse events; (ii) treatment outcome parameters were not improved by combining nivolumab with nab-sirolimus; and (iii) best responders were patients with undifferentiated pleomorphic sarcoma with PTEN loss and TSC2 mutation and estrogen receptor-positive leiomyosarcoma. Future direction in sarcoma research with nab-sirolimus will be biomarker-based (TSC1/2/mTOR, tumor mutational burden/mismatch repair deficiency etc.).


Subject(s)
Leiomyosarcoma , Sarcoma , Humans , Nivolumab/adverse effects , Leiomyosarcoma/drug therapy , Receptors, Estrogen , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , Sarcoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Mutation , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
7.
J Clin Oncol ; 39(33): 3660-3670, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34637337

ABSTRACT

PURPOSE: Malignant perivascular epithelioid cell tumor (PEComa) is a rare aggressive sarcoma, with no approved treatment. To our knowledge, this phase II, single-arm, registration trial is the first prospective clinical trial in this disease, investigating the safety and efficacy of the mammalian target of rapamycin inhibitor nab-sirolimus (AMPECT, NCT02494570). PATIENTS AND METHODS: Patients with malignant PEComa were treated with nab-sirolimus 100 mg/m2 intravenously once weekly for 2 weeks in 3-week cycles. The primary end point was objective response rate evaluated by independent radiology review. Key secondary end points included duration of response, progression-free survival, and safety. A key exploratory end point was tumor biomarker analysis. RESULTS: Thirty-four patients were treated (safety evaluable), and 31 were evaluable for efficacy. The overall response rate was 39% (12 of 31; 95% CI, 22 to 58) with one complete and 11 partial responses, 52% (16 of 31) of patients had stable disease, and 10% (3 of 31) had progressive disease. Responses were of rapid onset (67% by week 6) and durable. Median duration of response was not reached after a median follow-up for response of 2.5 years, with 7 of 12 responders with treatment ongoing (range, 5.6-47.2+ months). Twenty-five of 31 patients had tumor mutation profiling: 8 of 9 (89%) patients with a TSC2 mutation achieved a confirmed response versus 2 of 16 (13%) without TSC2 mutation (P < .001). The median progression-free survival was 10.6 months (95% CI, 5.5 months to not reached), and the median overall survival was 40.8 months (95% CI, 22.2 months to not reached). Most treatment-related adverse events were grade 1 or 2 and were manageable for long-term treatment. No grade ≥ 4 treatment-related events occurred. CONCLUSION: nab-Sirolimus is active in patients with malignant PEComa. The response rate, durability of response, disease control rate, and safety profile support that nab-sirolimus represents an important new treatment option for this disease.


Subject(s)
Albumins/metabolism , Antibiotics, Antineoplastic/therapeutic use , Nanoparticles/administration & dosage , Perivascular Epithelioid Cell Neoplasms/drug therapy , Sirolimus/therapeutic use , Adult , Aged , Albumins/chemistry , Antibiotics, Antineoplastic/chemistry , Female , Follow-Up Studies , Humans , Male , Middle Aged , Nanoparticles/chemistry , Neoplasm Metastasis , Perivascular Epithelioid Cell Neoplasms/pathology , Prognosis , Prospective Studies , Sirolimus/chemistry , Survival Rate
8.
Mol Clin Oncol ; 15(3): 186, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34277005

ABSTRACT

DeltaRex-G is a replication-incompetent amphotropic murine leukemia virus-based retroviral vector that displays a collagen-matrix-targeting decapeptide on its surface envelope protein, gp70, and encodes a cytocidal 'dominant negative', i.e. a truncated construct of the executive cyclin G1 (CCNG1) oncogene. DeltaRex-G inhibits the CCNG1 function of promoting cell competence and survival through the commanding CCNG1/cyclin-dependent kinase (CDK)/Myc/mouse double minute 2 homolog (Mdm2)/p53 axis. In 2009, DeltaRex-G was granted Fast Track designation from the US Food and Drug Administration for the treatment of pancreatic cancer. In 2019, the results of a phase 1/2 study that used DeltaRex-G as monotherapy for stage 4 chemotherapy-resistant pancreatic ductal adenocarcinoma (PDAC) were published. A unique participant of the aforementioned phase 1/2 study is now an 84-year-old Caucasian woman with chemoresistant PDAC who was treated with DeltaRex-G, 3x1011 colony forming units (cfu)/dose, 3 times/week for 4 weeks with a 2-week rest period, for 1.5 years. During the treatment period, the patient's tumors in the liver, lymph node and peritoneum exhibited progressive decreases in size, which were accompanied by a reduction and normalization of serum carbohydrate antigen 19-9 levels, and the patient achieved complete remission after 8 months of DeltaRex-G therapy with minimal side effects (grade 2 fatigue). Henceforth, the patient has been in remission for 12 years with no evidence of disease, no late therapy-related adverse events, and no further cancer therapy following DeltaRex-G treatment. The present study reports a mutation of tumor protein p53 (TP53) (G199V) found retrospectively in the patient's archived tumor samples. TP53 is a well-characterized tumor suppressor gene, and a critical regulatory component of the executive CCNG1/CDK/Myc/Mdm2/p53 axis, which regulates proliferative cell competence, DNA fidelity and survival. Studies are underway to determine whether TP53 mutations in pancreatic cancer can help identify a subset of patients with advanced metastatic cancer with an otherwise poor prognosis who would respond favorably to DeltaRex-G, which would broaden the treatment options for patients with otherwise lethal PDAC.

9.
Mol Ther Oncolytics ; 12: 56-67, 2019 Mar 29.
Article in English | MEDLINE | ID: mdl-30705966

ABSTRACT

Rexin-G is a replication-incompetent retroviral vector displaying a cryptic SIG-binding peptide for targeting abnormal Signature (SIG) proteins in tumors and encoding a dominant-negative human cyclin G1 construct. Herein we report on the safety and antitumor activity of escalating doses of Rexin-G in gemcitabine-refractory pancreatic adenocarcinoma, with one 10-year survivor. For the safety analysis (n = 20), treatment-related grade 1 adverse events included fatigue (n = 6), chills (n = 2), and headache (n = 1), with no organ damage and no DLT. No patient tested positive for vector-neutralizing antibodies, antibodies to gp70, replication-competent retrovirus (RCR), or vector integration into genomic DNA of peripheral blood lymphocytes (PBLs). For the efficacy analysis (n = 15), one patient achieved a complete response (CR), two patients had a partial response (PR), and 12 had stable disease (SD). Median progression-free survival (PFS) was 2.7, 4.0, and 5.6 months at doses 0-I, II, and III, respectively. Median overall survival (OS) and 1-year OS rate at dose 0-I were 4.3 months and 0%, and at dose II-III they were 9.2 months and 33.3%. To date, one patient is still alive with no evidence of cancer 10 years after the start of Rexin-G treatment. Taken together, these data suggest that Rexin-G, the first targeted gene delivery system, is uniquely safe and exhibits significant antitumor activity, for which the FDA granted fast-track designation.

11.
Mol Clin Oncol ; 9(2): 115-134, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30101008

ABSTRACT

Basic research in genetics, biochemistry and cell biology has identified the executive enzymes and protein kinase activities that regulate the cell division cycle of all eukaryotic organisms, thereby elucidating the importance of site-specific protein phosphorylation events that govern cell cycle progression. Research in cancer genomics and virology has provided meaningful links to mammalian checkpoint control elements with the characterization of growth-promoting proto-oncogenes encoding c-Myc, Mdm2, cyclins A, D1 and G1, and opposing tumor suppressor proteins, such as p53, pRb, p16INK4A and p21WAF1, which are commonly dysregulated in cancer. While progress has been made in identifying numerous enzymes and molecular interactions associated with cell cycle checkpoint control, the marked complexity, particularly the functional redundancy, of these cell cycle control enzymes in mammalian systems, presents a major challenge in discerning an optimal locus for therapeutic intervention in the clinical management of cancer. Recent advances in genetic engineering, functional genomics and clinical oncology converged in identifying cyclin G1 (CCNG1 gene) as a pivotal component of a commanding cyclin G1/Mdm2/p53 axis and a strategic locus for re-establishing cell cycle control by means of therapeutic gene transfer. The purpose of the present study is to provide a focused review of cycle checkpoint control as a practicum for clinical oncologists with an interest in applied molecular medicine. The aim is to present a unifying model that: i) clarifies the function of cyclin G1 in establishing proliferative competence, overriding p53 checkpoints and advancing cell cycle progression; ii) is supported by studies of inhibitory microRNAs linking CCNG1 expression to the mechanisms of carcinogenesis and viral subversion; and iii) provides a mechanistic basis for understanding the broad-spectrum anticancer activity and single-agent efficacy observed with dominant-negative cyclin G1, whose cytocidal mechanism of action triggers programmed cell death. Clinically, the utility of companion diagnostics for cyclin G1 pathways is anticipated in the staging, prognosis and treatment of cancers, including the potential for rational combinatorial therapies.

12.
Mol Clin Oncol ; 6(6): 861-865, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28588778

ABSTRACT

Soft tissue sarcoma is a rare neoplasm of mesenchymal origin, accounting for only ~1% of all adult cancers and consisting of 75 histological subtypes. In the present report, the unique case of a 14 year-old female with metastatic malignant peripheral nerve sheath tumor (formerly, malignant melanotic schwannoma) of the parotid gland, who experienced a durable response and sustained tumor control with Rexin-G®, a tumor-targeted retroviral expression vector encoding an anti-cyclin G1 construct, is described. Post-parotidectomy, and prior to the administration of Rexin-G®, the patient received various chemotherapy regimens, including doxorubicin, ifosfamide, temozolomide, sorafenib, and an immunological therapy with interleukin-2, which only resulted in the further progression of lung metastases. The patient subsequently participated in a Phase 1/2 gene therapy study, during which she received intravenous Rexin-G® as monotherapy for two years with minimal drug-associated adverse events. Currently, the patient has no evidence of active disease 9 years after commencing the Rexin-G® treatment, and with no additional anti-cancer therapy. In conclusion, Rexin-G® may be a viable therapeutic option for malignant peripheral nerve sheath tumors, and should be further investigated in prospective histology-specific clinical trials for this type, and possibly other types, of chemotherapy-resistant sarcoma.

13.
Adv Ther ; 33(7): 1055-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27234989

ABSTRACT

UNLABELLED: Trabectedin (ET743, Yondelis(®), manufactured by Baxter Oncology GmbH, Halle/Westfalen, Germany, for Janssen Products, LP, Horsham, PA), derived from the marine ascidian, Ecteinascidia turbinata, is a natural alkaloid with multiple complex mechanisms of action. On 23 October 2015, 15 years after the results of the first Phase 1 clinical trial using trabectedin for chemotherapy-resistant solid malignancies was reported, and 8 years after its approval in Europe, the United States Food and Drug Administration (USFDA) finally approved trabectedin for the treatment of unresectable or metastatic liposarcoma or leiomyosarcoma that has failed a prior anthracycline-containing regimen. Approval was based on the results of a pivotal Phase 3 trial involving a 2:1 randomization of 518 patients (who were further stratified by soft tissue sarcoma subtype), in which a significant improvement in progression-free survival was reported in the trabectedin-treated group vs. the dacarbazine-treated group (p < 0.001). In this trial, the most common adverse reactions were nausea, fatigue, vomiting, constipation, anorexia, diarrhea, peripheral edema, dyspnea, and headache, while the most serious were neutropenic sepsis, rhabdomyolysis, cardiomyopathy, hepatotoxicity, and extravasation leading to tissue necrosis. The most common grade 3-4 adverse events were laboratory abnormalities of myelosuppression in both arms and transient transaminitis in the trabectedin arm. In a recent Phase 2 trial, trabectedin had a similar outcome as doxorubicin when given as a single agent in the first-line setting. Studies are also being conducted to expand the use of trabectedin not only as a first-line cancer drug, but also for a number of other clinical indications, for example, in the case of mesenchymal chondrosarcoma, for which trabectedin has been reported to be exceptionally active. The possibility of combining trabectedin with targeted therapies, immune checkpoint inhibitors or virotherapy would also be an interesting concept. In short, trabectedin is an old new drug with proven potential to impact the lives of patients with soft tissue sarcoma and other solid malignancies. FUNDING: Sarcoma Oncology Center, Santa Monica, CA 90405.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dioxoles/therapeutic use , Sarcoma/drug therapy , Tetrahydroisoquinolines/therapeutic use , Antineoplastic Agents, Alkylating/adverse effects , Dioxoles/adverse effects , Disease-Free Survival , Europe , Germany , Humans , Neutropenia/chemically induced , Randomized Controlled Trials as Topic , Tetrahydroisoquinolines/adverse effects , Trabectedin
14.
Oncol Rep ; 24(4): 829-33, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20811660

ABSTRACT

The advent of pathotropic (disease-seeking) targeting has transported genetic medicine across the threshold of history with the progressive clinical validation of Rexin-G, a tumor-targeted nanosized anti-cancer agent. Achieving noteworthy single-agent efficacy and survival benefits in otherwise intractable cancers, the molecular biotechnology platform has stimulated intense interest in the underlying mechanisms-of-action. This report exhibits the effective localization of Rexin-G nanoparticles within a metastatic liver lesion, as observed upon its surgical excision.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Cyclin G1/genetics , Genetic Therapy/methods , Liver Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/secondary , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Genetic Vectors , Humans , Liver Neoplasms/secondary , Nanoparticles/therapeutic use , Pancreatic Neoplasms/pathology , Plasmids , Retroviridae
15.
Int J Oncol ; 36(6): 1341-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428757

ABSTRACT

The advent of pathotropic (disease-seeking) targeting technology has ushered cancer gene therapy across the threshold of history, marking the beginning of a new epoch of medical praxis. For the first time, clinical oncologists can reach beyond the finest of catheters, beyond the reach of the most gifted surgeons, to the very fabric of metastatic disease in an effort to halt the progression and turn the tide of otherwise intractable cancers. The enabling molecular biotechnologies embodied in the leading tumor-targeted agent, Rexin-G, and its timely development as a safe and effective anti-cancer drug - from oncogene discovery and target validation, to molecular engineering of the core nanotechnologies, to the first clinical proofs-of principle, confirmatory trials, expanded access programs, and accelerated regulatory approvals - have been extensively documented in the medical literature. Therefore, this paper represents a final chapter, highlighting a series of noteworthy cases studies in the emergent field of targeted genetic medicine: case studies which, in and of themselves, reveal vital and important aspects of the molecular-genetic bio-pharmacology, advanced clinical protocols, refinement of patient monitoring, expanding treatment options, and strategic medical approaches to patient care that exemplify and thereby extend the established principles of pathotropic targeting and cancer gene therapy to a new generation of clinical practitioners.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents/therapeutic use , Genetic Therapy/methods , Osteosarcoma/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Bone Neoplasms/therapy , Breast Neoplasms/therapy , Cyclin G1/genetics , Female , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Humans , Male , Middle Aged , Nanoparticles/therapeutic use , Ovarian Neoplasms/therapy , Pancreatic Neoplasms/therapy , Prostatic Neoplasms/therapy , Retroviridae/genetics , Sarcoma, Ewing/therapy
16.
Expert Opin Biol Ther ; 10(5): 819-32, 2010 May.
Article in English | MEDLINE | ID: mdl-20384524

ABSTRACT

IMPORTANCE OF THE FIELD: Rexin-G, a tumor-targeted retrovector bearing a cytocidal cyclin G1 construct, is the first targeted gene therapy vector to gain fast track designation and orphan drug priorities for multiple cancer indications in the US. AREAS COVERED IN THIS REVIEW: This review describes the major milestones in the clinical development of Rexin-G: from the molecular cloning and characterization of the human cyclin G1 proto-oncogene in 1994, to the design of the first knockout constructs and genetic engineering of the targeted delivery system from 1995 to 1997, through the initial proofs-of-concept, molecular pharmacology and toxicology studies of Rexin-G in preclinical cancer models from 1997 to 2001, to the pioneering clinical studies in humans from 2002 to 2004, which--together with the advancements in bioprocess development of high-potency clinical grade vectors circa 2005 - 2006--led to the accelerated approval of Rexin-G for all solid tumors by the Philippine FDA in 2007 and the rapid progression of clinical studies from 2007 to 2009 to the cusp of pivotal Phase III trials in the US. WHAT THE READER WILL GAIN: In recording the development of Rexin-G as a novel form of targeted biological therapy, this review also highlights important aspects of vector design engineering which served to overcome the physiological barriers to gene delivery as it addresses the key regulatory issues involved in the development of a targeted gene therapy product. TAKE HOME MESSAGE: Progressive clinical development of Rexin-G demonstrates the potential safety and efficacy of targeted genetic medicine, while validating the design engineering of the molecular biotechnology platform.


Subject(s)
Cyclin G1/genetics , Genetic Therapy/methods , Genetic Vectors , Neoplasms/therapy , Animals , Evidence-Based Medicine , Genetic Therapy/adverse effects , Humans , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Proto-Oncogene Mas , Treatment Outcome
17.
Mol Ther ; 18(2): 435-41, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19826403

ABSTRACT

Rexin-G, a nonreplicative pathology-targeted retroviral vector bearing a cytocidal cyclin G1 construct, was tested in a phase I/II study for gemcitabine-resistant pancreatic cancer. The patients received escalating doses of Rexin-G intravenously from 1 x 10(11) colony-forming units (cfu) 2-3x a week (dose 0-1) to 2 x 10(11) cfu 3x a week (dose 2) for 4 weeks. Treatment was continued if there was less than or equal to grade 1 toxicity. No dose-limiting toxicity (DLT) was observed, and no vector DNA integration, replication-competent retrovirus (RCR), or vector-neutralizing antibodies were noted. In nine evaluable patients, 3/3 patients had stable disease (SD) at dose 0-1. At dose 2, 1/6 patients had a partial response (PR) and 5/6 patients had SD. Median progression-free survival (PFS) was 3 months at dose 0-1, and >7.65 months at dose 2. Median overall survival (OS) was 4.3 months at dose 0-1, and 9.2 months at dose 2. One-year survival was 0% at dose 0-1 compared to 28.6% at dose 2, suggesting a dose-response relationship between OS and Rexin-G dosage. Taken together, these data indicate that (i) Rexin-G is safe and well tolerated, and (ii) Rexin-G may help control tumor growth, and may possibly prolong survival in gemcitabine-resistant pancreatic cancer, thus, earning US Food and Drug Administration's (FDA) fast-track designation as second-line treatment for pancreatic cancer.


Subject(s)
Cyclin G1/physiology , Pancreatic Neoplasms/therapy , Cyclin G1/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Genetic Therapy , Genetic Vectors/genetics , Humans , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/therapy , Pancreatic Neoplasms/drug therapy , Retroviridae/genetics , Gemcitabine
18.
Int J Oncol ; 35(2): 229-38, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578735

ABSTRACT

In an age where we can i) know precisely where a misplaced automobile resides by its global positioning, ii) send mechanistic probes to Mars with pinpoint accuracy, iii) calculate exactly how many mutations are required to create (i.e., to transform) a cancer cell, and iv) determine how many fewer genes it takes to develop a human being than it does a rice plant, it is difficult to fathom the previously unanswered question: 'Whatever happened to the promise and potential of cancer gene therapy?' This review answers that question with a resounding clinical dénouement. In addition, it provides a 'Cooks tour' of applied molecular genetics and nanotechnology as these fields relate to the development of Rexin-G the world's first tumor-targeted genetic medicine to be fully validated in the clinic. The commentary will expose certain fallacies and ideologies that have retarded the progress of cancer gene therapy as it advances our instruments and understanding of the finespun fabric of our nature.


Subject(s)
Cyclins/genetics , Genetic Therapy/methods , Genetic Vectors , Neoplasms/therapy , Retroviridae/genetics , Cell Proliferation , Cyclin G , Cyclin G1 , Humans , Injections , Nanotechnology , Neoplasms/pathology , Time Factors
19.
Mol Ther ; 17(9): 1651-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19532136

ABSTRACT

Rexin-G, a pathotropic nanoparticle bearing a cytocidal cyclin G1 construct was tested in a phase I/II study for chemotherapy-resistant sarcomas and a phase II study for chemotherapy-resistant osteosarcoma. Twenty sarcoma patients and 22 osteosarcoma patients received escalating doses of Rexin-G intravenously from 8 x 10(11) to 24 x 10(11) colony forming units (cfu)/cycle. Treatment was continued if there was or=3 months and median OS, 6.9 months. These studies suggest that Rexin-G is safe, may help control tumor growth, and may possibly improve survival in chemotherapy-resistant sarcoma and osteosarcoma.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Osteosarcoma/therapy , Retroviridae/genetics , Sarcoma/therapy , Adolescent , Adult , Aged , Cyclin G , Cyclin G1 , Cyclins/genetics , Cyclins/physiology , Drug Resistance, Neoplasm , Female , Genetic Therapy/adverse effects , Humans , Male , Middle Aged , Young Adult
20.
Int J Oncol ; 33(4): 665-75, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18813779

ABSTRACT

The advent of pathotropic (disease-seeking) targeting technologies, combined with advanced gene delivery vectors, provides a unique opportunity for the systemic delivery of immunomodulatory cytokine genes to remote sites of cancer metastasis. When injected intravenously, such pathotropic nanoparticles seek out and accumulate selectively at sites of tumor invasion and neo-angiogenesis, resulting in enhanced gene delivery, and thus cytokine production, within the tumor nodules. Used in conjunction with a primary tumoricidal agent (e.g., Rexin-G) that exposes tumor neoantigens, the tumor-targeted immunotherapy vector is intended to promote the recruitment and activation of host immune cells into the metastastic site(s), thereby initiating cancer immunization in situ. In this study, we examine the feasibility of cytokine gene delivery to cancerous lesions in vivo using intravenously administered pathotropically targeted nanoparticles bearing the gene encoding granulocyte/macrophage colony-stimulating factor (GM-CSF; i.e., Reximmune-C). In vitro, transduction of target cancer cells with Reximmune-C resulted in the quantitative production of bioactive and immunoreactive GM-CSF protein. In tumor-bearing nude mice, intravenous infusions of Reximmune-C-induced GM-CSF production by transduced cancer cells and paracrine secretion of the cytokine within the tumor nodules, which promoted the recruitment of host mononuclear cells, including CD40+ B cells and CD86+ dendritic cells, into the tumors. With the first proofs of principle established in preclinical studies, we generated an optimized vector configuration for use in advanced clinical trial designs, and extended the feasibility studies to the clinic. Targeted delivery and localized expression of the GM-CSF transgene was confirmed in a patient with metastatic cancer, as was the recruitment of significant tumor-infiltrating lymphocytes (TILs). Taken together, these studies provide the first demonstrations of cytokine gene delivery to cancerous lesions following intravenous administration and extend the applications of cancer immunization in vivo.


Subject(s)
Genetic Therapy/methods , Neoplasms/genetics , Neoplasms/therapy , Animals , Cancer Vaccines , Cell Line, Tumor , Gene Transfer Techniques , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immune System , Immunotherapy/methods , Mice , NIH 3T3 Cells , Neoplasm Metastasis , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...