23 results on '"Talimogene laherparepvec"'
Search Results
2. Locoregional melanoma metastases resistant to PD-1 inhibitor therapy treated with intralesional talimogene laherparepvec
- Author
-
Matthew M. Wallace, MD and John A. Zitelli, MD
- Subjects
melanoma ,Mohs surgery ,talimogene laherparepvec ,Dermatology ,RL1-803 - Published
- 2024
- Full Text
- View/download PDF
3. Liver transplant patient with in-transit squamous cell carcinoma treated with talimogene laherparepvec
- Author
-
Jamie Lebhar, BME, Jennifer Jacobs, BS, Khushnood Faraz, BS, April K.S. Salama, MD, and Paul J. Mosca, MD, PhD, MBA
- Subjects
intralesional ,in-transit ,liver transplant ,squamous cell carcinoma ,talimogene Laherparepvec ,T-VEC ,Dermatology ,RL1-803 - Published
- 2023
- Full Text
- View/download PDF
4. Use of Talimogene Laherparepvec to Treat Cutaneous Squamous Cell Carcinoma in a Renal Transplant Patient
- Author
-
Max Miller, Nancy H Kim, Maya K Thosani, and Justin C Moser
- Subjects
transplant associated squamous cell carcinoma ,talimogene laherparepvec ,case report ,immunotherapy ,organ transplant recipient ,Dermatology ,RL1-803 - Abstract
A 66-year-old female with a history of two renal transplants due to recurrent thrombotic thrombocytopenic purpura presented to clinic with multiple lesions identified to be non-metastatic cutaneous squamous cell carcinoma (CSCC). The patient previously underwent multiple Mohs procedures and radiation therapy treatment but continued to develop CSCC lesions with increasing frequency. After discussing multiple treatment options, it was elected to pursue treatment with Talimogene laherparepvec (T-VEC) given the systemic immune responses it can cause, with low theoretical risk of graft rejection. After starting intratumoral T-VEC injections, treated lesions began to decrease in size, and a reduction in the rate of new CSCC lesions was observed. Treatment was held due to unrelated renal complications during which time new CSCCs developed. Patient was restarted on T-VEC therapy with no recurrent renal issues. Upon reinitiating treatment, injected and non-injected lesions showed reduction in size, and the development of new lesions again ceased. One injected lesion was resected via Mohs micrographic surgery due to its size and discomfort. On sectioning, this demonstrated an exuberant lymphocytic perivascular infiltrate which was consistent with treatment response to T-VEC, with little active tumor. With high rates of non-melanoma skin cancer in renal transplant patients, their transplant status significantly limits treatment options, specifically with regards to anti-PD-1 therapy. This case suggests T-VEC can generate local and systemic immune responses in the setting of immunosuppression and that T-VEC may be a beneficial therapeutic option for transplant patients with CSCC.
- Published
- 2023
- Full Text
- View/download PDF
5. Multimodal Therapy Approaches for NUT Carcinoma by Dual Combination of Oncolytic Virus Talimogene Laherparepvec with Small Molecule Inhibitors
- Author
-
Stavros Sotiriadis, Julia Beil, Susanne Berchtold, Irina Smirnow, Andrea Schenk, and Ulrich M. Lauer
- Subjects
NUT carcinoma ,talimogene laherparepvec ,T-VEC ,virotherapy ,small molecule inhibitors ,combination therapy ,Microbiology ,QR1-502 - Abstract
NUT (nuclear-protein-in-testis) carcinoma (NC) is a highly aggressive tumor disease. Given that current treatment regimens offer a median survival of six months only, it is likely that this type of tumor requires an extended multimodal treatment approach to improve prognosis. In an earlier case report, we could show that an oncolytic herpes simplex virus (T-VEC) is functional in NC patients. To identify further combination partners for T-VEC, we have investigated the anti-tumoral effects of T-VEC and five different small molecule inhibitors (SMIs) alone and in combination in human NC cell lines. Dual combinations were found to result in higher rates of tumor cell reductions when compared to the respective monotherapy as demonstrated by viability assays and real-time tumor cell growth monitoring. Interestingly, we found that the combination of T-VEC with SMIs resulted in both stronger and earlier reductions in the expression of c-Myc, a main driver of NC cell proliferation, when compared to T-VEC monotherapy. These results indicate the great potential of combinatorial therapies using oncolytic viruses and SMIs to control the highly aggressive behavior of NC cancers and probably will pave the way for innovative multimodal clinical studies in the near future.
- Published
- 2024
- Full Text
- View/download PDF
6. Neoadjuvant nivolumab + T-VEC combination therapy for resectable early stage or metastatic (IIIB-IVM1a) melanoma with injectable disease: study protocol of the NIVEC trial
- Author
-
Maartje W. Rohaan, Emma H. A. Stahlie, Viola Franke, Lisanne P. Zijlker, Sofie Wilgenhof, Vincent van der Noort, Alexander C. J. van Akkooi, and John B. A. G. Haanen
- Subjects
Melanoma ,Early metastatic ,Neoadjuvant ,Talimogene laherparepvec ,Nivolumab ,Immune checkpoint inhibitor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Trials investigating neoadjuvant treatment with immune checkpoint inhibitors (ICI) in patients with melanoma have shown high clinical and pathologic response rates. Treatment with talimogene laherparepvec (T-VEC), a modified herpes simplex virus type-1 (HSV-1), is approved for patients with unresectable stage IIIB-IVM1a melanoma and has the potential to make tumors more susceptible for ICI. Combination ICI and intralesional T-VEC has already been investigated in patients with unresectable stage IIIB-IV disease, however, no data is available yet on the potential benefit of this combination therapy in neoadjuvant setting. Methods This single center, single arm, phase II study aims to show an improved major pathologic complete response (pCR) rate, either pCR or near-pCR, up to 45% in 24 patients with resectable stage IIIB-IVM1a melanoma upon neoadjuvant combination treatment with intralesional T-VEC and systemic nivolumab (anti-PD-1 antibody). Patients will receive four courses of T-VEC up to 4 mL (first dose as seroconversion dose) and three doses of nivolumab (240 mg flatdose) every 2 weeks, followed by surgical resection in week nine. The primary endpoint of this trial is pathologic response rate. Secondary endpoints are safety, the rate of delay of surgery and event-free survival. Additionally, prognostic and predictive biomarker research and health-related quality of life evaluation will be performed. Discussion Intralesional T-VEC has the capacity to heighten the immune response and to elicit an abscopal effect in melanoma in combination with ICI. However, the potential clinical benefit of T-VEC plus ICI in the neoadjuvant setting remains unknown. This is the first trial investigating the efficacy and safety of neoadjuvant treatment of T-VEC and nivolumab followed by surgical resection in patients with stage IIIB-IVM1a melanoma, with the potential of high pathologic response rates and acceptable toxicity. Trial registration This trial was registered in the European Union Drug Regulating Authorities Clinical Trials Database (EudraCT- number: 2019–001911-22 ) and the Central Committee on Research Involving Human Subjects (NL71866.000.19) on 4th June 2020. Secondary identifying number: NCT04330430 .
- Published
- 2022
- Full Text
- View/download PDF
7. A phase 1, first-in-child, multicenter study to evaluate the safety and efficacy of the oncolytic herpes virus talimogene laherparepvec in pediatric patients with advanced solid tumors
- Author
-
Lucas Moreno, Pierre Teira, James M. Croop, Nicolas U. Gerber, Nicolas André, Isabelle Aerts, Luis Gros Subias, Bram De Wilde, Francisco Bautista, Brian Turpin, Srinivasa Kunduri, Ali Hamidi, Tatiana Lawrence, and Keri A. Streby
- Subjects
immunotherapy ,pediatric solid tumor ,talimogene laherparepvec ,non-CNS tumors ,relapsed and refractory cancer ,oncolytic herpes virus ,Pediatrics ,RJ1-570 - Abstract
BackgroundThe survival rates for pediatric patients with relapsed and refractory tumors are poor. Successful treatment strategies are currently lacking and there remains an unmet need for novel therapies for these patients. We report here the results of a phase 1 study of talimogene laherparepvec (T-VEC) and explore the safety of this oncolytic immunotherapy for the treatment of pediatric patients with advanced non–central nervous system tumors.MethodsT-VEC was delivered by intralesional injection at 106 plaque-forming units (PFU)/ml on the first day, followed by 108 PFU/ml on the first day of week 4 and every 2 weeks thereafter. The primary objective was to evaluate the safety and tolerability as assessed by the incidence of dose-limiting toxicities (DLTs). Secondary objectives included efficacy indicated by response and survival per modified immune-related response criteria simulating the Response Evaluation Criteria in Solid Tumors (irRC-RECIST).ResultsFifteen patients were enrolled into two cohorts based on age: cohort A1 (n = 13) 12 to ≤21 years old (soft-tissue sarcoma, n = 7; bone sarcoma, n = 3; neuroblastoma, n = 1; nasopharyngeal carcinoma, n = 1; and melanoma, n = 1) and cohort B1 (n = 2) 2 to
- Published
- 2023
- Full Text
- View/download PDF
8. Activity of TNT: a phase 2 study using talimogene laherparepvec, nivolumab and trabectedin for previously treated patients with advanced sarcomas (NCT# 03886311)
- Author
-
Sant P. Chawla, Walter Andree Tellez, Hripsime Chomoyan, Chrysler Valencia, Amir Ahari, Nadezhda Omelchenko, Stefan Makrievski, Don A. Brigham, Victoria Chua-Alcala, Doris Quon, Ania Moradkhani, and Erlinda M. Gordon
- Subjects
talimogene laherparepvec ,nivolumab ,trabectedin ,sarcoma ,immunotherapy ,alkylating agents ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
BackgroundIntratumoral 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 methodsThis 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).ResultsMedian 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%).ConclusionsTaken 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.
- Published
- 2023
- Full Text
- View/download PDF
9. Durable melanoma control following disseminated talimogene laherparepvec herpetic infection
- Author
-
Leonid Shmuylovich, MD, PhD, Aubriana M. McEvoy, MD, MS, Ryan C. Fields, MD, Leonel Hernandez-Aya, MD, George Ansstas, MD, and David Y. Chen, MD, PhD
- Subjects
disseminated herpes infection ,durable response ,melanoma ,talimogene laherparepvec ,T-VEC ,Dermatology ,RL1-803 - Published
- 2022
- Full Text
- View/download PDF
10. Complete remission from intralesional talimogene laherparepvec for regionally advanced Merkel cell carcinoma in an immunocompromised solid organ transplant patient
- Author
-
Kelsey E. Hirotsu, MD, Vivian Hua, BA, Anhthy T. Tran, MSN, Laura Morris, BSN, Sunil A. Reddy, MD, Bernice Y. Kwong, MD, and Lisa C. Zaba, MD, PhD
- Subjects
cutaneous oncology ,herpes ,immunosuppressed ,Merkel cell carcinoma ,oncodermatology ,talimogene laherparepvec ,Dermatology ,RL1-803 - Published
- 2021
- Full Text
- View/download PDF
11. Unraveling the Effects of a Talimogene Laherparepvec (T-VEC)-Induced Tumor Oncolysate on Myeloid Dendritic Cells
- Author
-
Jens Tijtgat, Jolien De Munck, Inès Dufait, Julia Katharina Schwarze, Ivan Van Riet, Lorenzo Franceschini, Karine Breckpot, Joeri L. Aerts, Bart Neyns, and Sandra Tuyaerts
- Subjects
melanoma ,myeloid dendritic cell ,BDCA-1 ,BDCA-3 ,Talimogene laherparepvec ,cell therapy ,Immunologic diseases. Allergy ,RC581-607 - Abstract
T-VEC, a HSV-1 derived oncolytic virus, is approved for the treatment of advanced melanoma. The mechanisms that underly the systemic anti-tumor effect that is seen following intratumoral injection have not yet been studied but are likely to be mediated by myeloid dendritic cells (myDC) that initiate an adaptive immune response. In this study we could demonstrate that T-VEC is non-toxic for human myDC. T-VEC and a T-VEC oncolysate of melanoma cell lines were able to mature human myDC. myDC were able to take up lysed melanoma cells and cross-present melanoma-derived tumor antigens to antigen-specific T cells. Our results support the possible role of myDC as mediators of an adaptive anti-tumor effect and intratumoral co-administration of T-VEC plus autologous myDC could be a complementary treatment option. A clinical trial that investigates this hypothesis is currently ongoing.
- Published
- 2021
- Full Text
- View/download PDF
12. Final analyses of OPTiM: a randomized phase III trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor in unresectable stage III–IV melanoma
- Author
-
Robert H. I. Andtbacka, Frances Collichio, Kevin J. Harrington, Mark R. Middleton, Gerald Downey, Katarina Ӧhrling, and Howard L. Kaufman
- Subjects
Efficacy ,Final analysis ,Melanoma ,Overall survival ,Talimogene laherparepvec ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Talimogene laherparepvec is an oncolytic immunotherapy approved in the US, Europe, Australia and Switzerland. We report the final planned analysis of OPTiM, a randomized open-label phase III trial in patients with unresectable stage IIIB–IVM1c melanoma. Methods Patients were randomized 2:1 to receive intratumoral talimogene laherparepvec or subcutaneous recombinant GM-CSF. In addition to overall survival (OS), durable response rate (DRR), objective response rate (ORR), complete responses (CR), and safety are also reported. All final analyses are considered to be descriptive and treatment responses were assessed by the investigators. Results Of 436 patients in the intent-to-treat population, 295 were allocated to talimogene laherparepvec and 141 to GM-CSF. Median follow-up in the final OS analysis was 49 months. Median OS was 23.3 months (95% confidence interval [CI], 19.5–29.6) and 18.9 months (95% CI, 16.0–23.7) in the talimogene laherparepvec and GM-CSF arms, respectively (unstratified hazard ratio, 0.79; 95% CI, 0.62–1.00; p = 0.0494 [descriptive]). DRR was 19.0 and 1.4% (unadjusted odds ratio, 16.6; 95% CI, 4.0–69.2; p
- Published
- 2019
- Full Text
- View/download PDF
13. Observational study of talimogene laherparepvec use in the anti-PD-1 era for melanoma in the US (COSMUS-2)
- Author
-
James Sun, Brian R Gastman, Lucy McCahon, Elizabeth I Buchbinder, Igor Puzanov, Michele Nanni, James M Lewis, Richard D Carvajal, Shahnaz Singh-Kandah, Anupam M Desai, Leon Raskin, Carrie M Nielson, Rubina Ismail, and Jonathan S Zager
- Subjects
combination therapy ,immunotherapy ,metastatic melanoma ,oncolytic virus ,real-world evidence ,talimogene laherparepvec ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Aim: Talimogene laherparepvec (T-VEC) is an intralesional therapy for unresectable, metastatic melanoma. T-VEC real-world use in the context of anti-PD1-based therapy requires further characterization. Materials & methods: A retrospective review of T-VEC use from 1 January 2017 and 31 March 2018 for melanoma patients was conducted at seven US institutions. Results: Among 83 patients, three categories of T-VEC and anti-PD-1 therapy were identified: T-VEC used without anti-PD-1 (n = 29, 35%), T-VEC after anti-PD-1-based therapy (n = 22, 27%) and concurrent T-VEC and anti-PD-1-based therapy (n = 32, 39%). 25% of patients discontinued T-VEC therapy due to no remaining injectable lesions, 37% discontinued T-VEC due to progressive disease. Discontinuation of T-VEC did not differ by anti-PD-1-based therapy use or timing. Conclusion: In real-world settings, T-VEC may be used concurrently with or after anti-PD-1-based therapy.
- Published
- 2020
- Full Text
- View/download PDF
14. Talimogene Laherparepvec in Advanced Mucosal Melanoma of the Urethra Upon Primary Resistance on Immune Checkpoint Inhibition: A Case Report
- Author
-
Anne Fröhlich, Friederike Hoffmann, Dennis Niebel, Eva Egger, Guido M. Kukuk, Marieta Toma, Judith Sirokay, Thomas Bieber, and Jennifer Landsberg
- Subjects
immune checkpoint blockade ,intralesional treatment ,mucosal melanoma ,primary resistance ,talimogene laherparepvec ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Mucosal melanomas including melanomas of the urogenital tract represent a rare type of melanoma characterized by low mutational burden and poor prognosis. Immune checkpoint inhibition has so far only been assessed in a limited number of mucosal melanoma patients and, in contrast to response in cutaneous melanoma, was associated with disappointing response rates. The oncolytic viral immunotherapy Talimogene laherparepvec (T-VEC) has recently been approved for treatment of locally advanced or unresectable melanoma. T-VEC combines direct oncolytic effects with local and systemic immune-mediated anti-tumor response. Our rationale to use T-VEC in this case was an expected augmentation of immunogenicity by tumor lysis to overcome primary resistance of a mucosal melanoma to immune checkpoint blockade.Objective: To report the first case of an advanced mucosal melanoma of the urethra treated with intralesional application of Talimogene laherparepvec.Case Report: A 78-years old female patient was diagnosed with an advanced mucosal melanoma of the urethra with inguinal lymph node metastases and intravaginal mucosal metastases. Shortly after surgical resection of the tumor mass, intravaginal mucosal metastases, and new nodal metastases in proximity of the left iliac vessels were diagnosed. The patient was treated with the anti-PD1 antibody pembrolizumab and obtained a stable disease lasting for 30 weeks. However, upon checkpoint inhibition the patient developed a loco-regional progressive disease featuring bleeding intravaginal metastases, while nodal metastases remained stable. We stopped treatment with pembrolizumab and administered T-VEC directly into the intravaginal mucosal metastases. After five injections T-VEC yielded a partial response with clinical regression of the injected mucosal metastases. Disease remained stable for 16 weeks under biweekly T-VEC treatment. Thereafter the patient showed disease progression in nodal metastases. T-VEC was discontinued. Immunotherapy with pembrolizumab was restarted but failed to achieve a response. Finally, targeted therapy with imatinib was induced in presence of a druggable c-KIT mutation, leading to a considerable response of all tumor sites that is still ongoing.Conclusion: T-VEC represents an effective and well-tolerated treatment option for patients with loco-regionally advanced mucosal melanoma. In combination with immunotherapy, T-VEC bears the potential of synergistic effects to overcome the specific primary resistance of mucosal melanoma to immune checkpoint blockade.
- Published
- 2020
- Full Text
- View/download PDF
15. Kaposi’s varicelliform eruption in a patient with metastatic melanoma and primary cutaneous anaplastic large cell lymphoma treated with talimogene laherparepvec and nivolumab
- Author
-
David M. Miller, Ryan M. Trowbridge, Anupam Desai, and Reed E. Drews
- Subjects
Talimogene laherparepvec ,Melanoma ,Anaplastic large cell lymphoma ,Kaposi varicelliform eruption ,eczema TVECium ,Immunocompromised ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Immune-directed therapies have become front-line therapy for melanoma and are transforming the management of advanced disease. In refractory cases, multi-modal immunoncology (IO) approaches are being utilized, including combining immune checkpoint blockade (ICB) with oncolytic herpes viruses. Talimogene laherparepvec (T-VEC) is the first genetically modified oncolytic viral therapy (OVT) approved for the treatment of recurrent and unresectable melanoma. The use of IO in patients with concomitant malignancies and/or compromised immune systems is limited due to systematic exclusion from clinical trials. For example, a single case report of a solid organ transplant patient successfully treated with T-VEC for metastatic melanoma has been reported. Furthermore, the use of ICB in T-cell malignancies is limited and paradoxical worsening has been described. To our knowledge, this is the first report of dual ICB/T-VEC being administered to a patient with concurrent primary cutaneous anaplastic large cell lymphoma (pcALCL) and melanoma. Case presentation Here we present the case of a patient with concomitant primary cutaneous ALCL and metastatic melanoma, progressing on anti-programmed death (PD)-1 therapy, who developed Kaposi’s varicelliform eruption after receiving the first dose of Talimogene laherparepvec. Conclusion This case highlights the complexities of care of patients with coexistent cancers, demonstrates rapid progression of primary cutaneous ALCL on nivolumab and introduces a novel adverse effect of Talimogene laherparepvec.
- Published
- 2018
- Full Text
- View/download PDF
16. Talimogene laherparepvec: First in class oncolytic virotherapy
- Author
-
Robert M. Conry, Brian Westbrook, Svetlana McKee, and Timothy Graham Norwood
- Subjects
talimogene laherparepvec ,oncolytic virus ,melanoma ,herpes simplex viral type i ,granulocyte-macrophage colony-stimulating factor ,granulomatous dermatitis ,immune checkpoint blockade ,Immunologic diseases. Allergy ,RC581-607 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Oncolytic viruses represent a novel drug class in which native or modified viruses mediate tumor regression through selective replication within and lysis of tumor cells as well as induction of systemic antitumor immunity capable of eradicating tumor at distant, uninjected sites. Talimogene laherparepvec (TVEC) is a type I herpes simplex virus genetically modified to preferentially replicate in tumor cells, enhance antigen loading of MHC class I molecules and express granulocyte-macrophage colony-stimulating factor to increase tumor-antigen presentation by dendritic cells. It is presently the only oncolytic virus approved by the FDA with an indication for advanced melanoma based upon improved durable response rate in a randomized, phase III trial. Clinical trials are underway in melanoma investigating TVEC as neoadjuvant monotherapy and in combination with checkpoint inhibitors for unresectable disease as well as in an array of other malignancies. It is appropriate to review TVEC's biology mechanism of action, clinical indication and future directions as a prototype of the burgeoning class of oncolytic viruses.
- Published
- 2018
- Full Text
- View/download PDF
17. Complete intracranial response to talimogene laherparepvec (T-Vec), pembrolizumab and whole brain radiotherapy in a patient with melanoma brain metastases refractory to dual checkpoint-inhibition
- Author
-
Zoë Blake, Douglas K. Marks, Robyn D. Gartrell, Thomas Hart, Patti Horton, Simon K. Cheng, Bret Taback, Basil A. Horst, and Yvonne M. Saenger
- Subjects
Melanoma ,Brain metastases ,Talimogene laherparepvec ,T-Vec ,Checkpoint inhibitors ,Anti-PD1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Immunotherapy, in particular checkpoint blockade, has changed the clinical landscape of metastatic melanoma. Nonetheless, the majority of patients will either be primary refractory or progress over follow up. Management of patients progressing on first-line immunotherapy remains challenging. Expanded treatment options with combination immunotherapy has demonstrated efficacy in patients previously unresponsive to single agent or alternative combination therapy. Case presentation We describe the case of a patient with diffusely metastatic melanoma, including brain metastases, who, despite being treated with stereotactic radiosurgery and dual CTLA-4/PD-1 blockade (ipilimumab/nivolumab), developed systemic disease progression and innumerable brain metastases. This patient achieved a complete CNS response and partial systemic response with standard whole brain radiation therapy (WBRT) combined with Talimogene laherparepvec (T-Vec) and pembrolizumab. Conclusion Patients who do not respond to one immunotherapy combination may respond during treatment with an alternate combination, even in the presence of multiple brain metastases. Biomarkers are needed to assist clinicians in evidence based clinical decision making after progression on first line immunotherapy to determine whether response can be achieved with second line immunotherapy.
- Published
- 2018
- Full Text
- View/download PDF
18. Durable response rate as an endpoint in cancer immunotherapy: insights from oncolytic virus clinical trials
- Author
-
Howard L. Kaufman, Robert H. I. Andtbacka, Frances A. Collichio, Michael Wolf, Zhongyun Zhao, Mark Shilkrut, Igor Puzanov, and Merrick Ross
- Subjects
Melanoma ,Talimogene laherparepvec ,Durable response rate ,Patient-reported outcomes ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Traditional response criteria may be insufficient to characterize full clinical benefits of anticancer immunotherapies. Consequently, endpoints such as durable response rate (DRR; a continuous response [complete or partial objective response] beginning within 12 months of treatment and lasting ≥6 months) have been employed. There has not, however, been validation that DRR correlates with other more traditional endpoints of clinical benefit such as overall survival. Methods We evaluated whether DRR was associated with clinically meaningful measures of benefit (eg, overall survival [OS], quality of life [QoL], or treatment-free interval [TFI]) in a phase 3 clinical trial of an oncolytic virus for melanoma treatment. To evaluate the association between DRR and OS and to mitigate lead time bias, landmark analyses were used. QoL was evaluated using the FACT-BRM questionnaire (comprising the FACT-BRM Physical, Social/Family, Emotional, and Functional well-being domains, the Additional Concerns, Physical and Mental treatment-specific subscales, and the Trial Outcome Index [TOI]). TFI was defined as time from the last study therapy dose to first subsequent therapy dose (including any systemic anticancer therapy for melanoma after study therapy discontinuation). Results Four hundred thirty-six patients were included in the intent-to-treat population. Achieving DR was associated with a statistically significant improvement in OS in a landmark analysis at 9 months (HR = 0.07; P = 0.0003), 12 months (HR = 0.05, P
- Published
- 2017
- Full Text
- View/download PDF
19. Inflammatory melanoma in transit metastases with complete response to talimogene laherparepvec
- Author
-
Jonathan T. Blackmon, Michael S. Stratton, MD, Young Kwak, MD, Peter G. Pavlidakey, MD, Andrzej T. Slominski, MD, Svetlana B. McKee, RN, BSN, Toni M. Viator, RN, Ju Young Kim, PhD, Conway C. Huang, MD, and Robert M. Conry, MD
- Subjects
dermal intercalation ,dermal lymphatics ,erythematous plaque ,immune phenotype ,inflammatory melanoma ,talimogene laherparepvec ,Dermatology ,RL1-803 - Published
- 2017
- Full Text
- View/download PDF
20. Talimogene laherparepvec for regionally advanced Merkel cell carcinoma: A report of 2 cases
- Author
-
Jonathan T. Blackmon, Ratika Dhawan, MBBS, MPH, Toni M. Viator, RN, Nina L. Terry, MD, JD, and Robert M. Conry, MD
- Subjects
clinical response ,Merkel cell carcinoma ,oncolytic viral immunotherapy ,talimogene laherparepvec ,Dermatology ,RL1-803 - Published
- 2017
- Full Text
- View/download PDF
21. Inflammatory Colitis after Treatment of Melanoma with Talimogene Laherparepvec (T-VEC)
- Author
-
Jennifer Gardner, Teresa Hyun, Gideon Steinbach, and John Thompson
- Subjects
colitis ,immune-related adverse event ,immunotherapy ,melanoma ,talimogene laherparepvec ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Talimogene laherparepvec (T-VEC) is an intralesional oncolytic viral therapy for the treatment of recurrent, unresectable cutaneous, subcutaneous, and nodal melanoma. It is thought to work through direct tumor oncolysis and by eliciting a tumor-specific systemic immune response. Immune-related adverse events have been reported only rarely with this therapy. We report a case of culture-negative, biopsy-proven colitis following pathologic complete response of recurrent, and intransit cutaneous melanoma to T-VEC. To the best of our knowledge, this is the first report of immune-related colitis following T-VEC.
- Published
- 2018
- Full Text
- View/download PDF
22. Genomic Signature of the Natural Oncolytic Herpes Simplex Virus HF10 and Its Therapeutic Role in Preclinical and Clinical Trials
- Author
-
Ibrahim Ragab Eissa, Yoshinori Naoe, Itzel Bustos-Villalobos, Toru Ichinose, Maki Tanaka, Wu Zhiwen, Nobuaki Mukoyama, Taishi Morimoto, Noriyuki Miyajima, Hasegawa Hitoki, Seiji Sumigama, Branko Aleksic, Yasuhiro Kodera, and Hideki Kasuya
- Subjects
herpes simplex oncolytic viruses ,genomic structure ,HF10 ,talimogene laherparepvec ,preclinical studies ,combination therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Oncolytic viruses (OVs) are opening new possibilities in cancer therapy with their unique mechanism of selective replication within tumor cells and triggering of antitumor immune responses. HF10 is an oncolytic herpes simplex virus-1 with a unique genomic structure that has non-engineered deletions and insertions accompanied by frame-shift mutations, in contrast to the majority of engineered OVs. At the genetic level, HF10 naturally lacks the expression of UL43, UL49.5, UL55, UL56, and latency-associated transcripts, and overexpresses UL53 and UL54. In preclinical studies, HF10 replicated efficiently within tumor cells with extensive cytolytic effects and induced increased numbers of activated CD4+ and CD8+ T cells and natural killer cells within the tumor, leading to a significant reduction in tumor growth and prolonged survival rates. Investigator-initiated clinical studies of HF10 have been completed in recurrent breast carcinoma, head and neck cancer, and unresectable pancreatic cancer in Japan. Phase I trials were subsequently completed in refractory superficial cancers and melanoma in the United States. HF10 has been demonstrated to have a high safety margin with low frequency of adverse effects in all treated patients. Interestingly, HF10 antigens were detected in pancreatic carcinoma over 300 days after treatment with infiltration of CD4+ and CD8+ T cells, which enhanced the immune response. To date, preliminary results from a Phase II trial have indicated that HF10 in combination with ipilimumab (anti-CTLA-4) is safe and well tolerated, with high antitumor efficacy. Improvement of the effect of ipilimumab was observed in patients with stage IIIb, IIIc, or IV unresectable or metastatic melanoma. This review provides a concise description of the genomic functional organization of HF10 compared with talimogene laherparepvec. Furthermore, this review focuses on HF10 in cancer treatment as monotherapy as well as in combination therapy through a concise description of all preclinical and clinical data. In addition, we will address approaches for future directions in HF10 studies as cancer therapy.
- Published
- 2017
- Full Text
- View/download PDF
23. Oncolytic virotherapy as emerging immunotherapeutic modality: potential of parvovirus H-1
- Author
-
Markus eMoehler, Katrin eGoepfert, Bernd eHeinrich, Caroline eBreitbach, Maike eDelic, Peter Robert Galle, and Jean eRommelaere
- Subjects
Immunotherapy ,CTLA-4 ,Dentritic cells ,autonomous parvovirus ,H-1PV ,talimogene laherparepvec ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Human tumors develop multiple strategies to evade recognition and efficient suppression by the immune system. Therefore, a variety of immunotherapeutic strategies have been developed to reactivate and reorganize the human immune system. The recent development of new antibodies against immune check points may help to overcome the immune silencing induced by human tumors. Some of these antibodies have already been approved for treatment of various solid tumor entities. Interestingly, targeting antibodies may be combined with standard chemotherapy or radiation protocols. Furthermore, recent evidence indicates that intratumoral (it) or intravenous (iv) injections of replicative oncolytic viruses such as herpes simplex-, pox-, parvo- or adenoviruses may also reactivate the human immune system. By generating tumor cell lysates in situ, oncolytic viruses overcome cellular tumor resistance mechanisms and induce immunogenic tumor cell death resulting in the recognition of newly released tumor antigens.This is in particular the case of the oncolytic parvovirus H-1 (H-1PV) which is able to kill human tumor cells and stimulate an antitumor immune response through increased presentation of tumor-associated antigens, maturation of dendritic cells and release of proinflammatory cytokines. Current research and clinical studies aim to assess the potential of oncolytic virotherapy and its combination with immunotherapeutic agents or conventional treatments to further induce effective antitumoral immune responses.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.