14 results on '"Robert A. Dillman"'
Search Results
2. Randomized phase II trial of autologous dendritic cell vaccines versus autologous tumor cell vaccines in metastatic melanoma: 5-year follow up and additional analyses
- Author
-
Robert O. Dillman, Andrew N. Cornforth, Gabriel I. Nistor, Edward F. McClay, Thomas T. Amatruda, and Carol Depriest
- Subjects
Metastatic melanoma ,Patient-specific vaccines ,Dendritic-cell vaccines ,Tumor-cell vaccines ,Autologous tumor cell lines ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Despite improved survival following checkpoint inhibitors, there is still a potential role for anti-cancer therapeutic vaccines. Because of biological heterogeneity and neoantigens resulting from each patient’s mutanome, autologous tumor may be the best source of tumor-associated antigens (TAA) for vaccines. Ex vivo loading of autologous dendritic cells with TAA may be associated with superior clinical outcome compared to injecting irradiated autologous tumor cells. We conducted a randomized phase II trial to compare autologous tumor cell vaccines (TCV) and autologous dendritic cell vaccines (DCV) loaded with autologous TAA. Methods Short-term autologous tumor cell lines were established from metastatic tumor. Vaccines were admixed with 500 micrograms of GM-CSF and injected weekly for 3 weeks, then at weeks 8, 12,16, 20, and 24. The primary endpoint was overall survival. Secondary objectives were identification of adverse events, and results of delayed type hypersensitivity (DTH) reactions to intradermal tumor cell injections. Results Forty-two patients were randomized. All were followed from randomization until death or for five years; none were lost to follow-up. DCV was associated with longer survival: median 43.4 versus 20.5 months (95% CI, 18.6 to > 60 versus 9.3 to 32.3 months) and a 70% reduction in the risk of death (hazard ratio = 0.304, p = 0.0053, 95% CI, 0.131 to 0.702). Tumor DTH reactions were neither prognostic nor predictive. The most common treatment-related adverse events were mild to moderate local injection site reactions and flu-like symptoms; but grade 2 treatment-related adverse events were more frequent with TCV. Serum marker analyses at week-0 and week-4 showed that serum markers were similar at baseline in each arm, but differed after vaccination. Conclusions This is the only human clinical trial comparing DCV and TCV as platforms for autologous TAA presentation. DCV was associated with minimal toxicity and long-term survival in patients with metastatic melanoma. DTH to autologous tumor cells was neither prognostic for survival nor predictive of benefit for either vaccine. Trial registration Clinical trials.gov NCT00948480 retrospectively registered 28 July 2009.
- Published
- 2018
- Full Text
- View/download PDF
3. 952 Phase II trial of AV-GBM-1: dendritic cell vaccine pulsed with lysate enriched for autologous tumor-initiating cell antigens in the treatment of patients with newly diagnosed glioblastoma
- Author
-
Thomas H. Taylor, Renato V. LaRocca, Daniela A. Bota, Jose Carrillo, Mehrdad Abedi, Robert O. Dillman, David Piccioni, Xiao-Tang Kong, Candace Hsieh, Gabriel Nistor, Christopher Duma, Robert Aiken, Santosh Kesari, and Frank P.K. Hsu
- Subjects
Pharmacology ,Cancer Research ,Lysis ,business.industry ,Immunology ,Cell ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Newly diagnosed ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Antigen ,Dendritic cell vaccine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 ,Autologous tumor ,Glioblastoma - Abstract
BackgroundStandard therapy of glioblastoma (GBM), which includes maximum safe resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ, is associated with poor overall survival (OS). Adding treatment with AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve OS. A multi-center phase II clinical trial was conducted to determine feasibility, safety, and efficacy of AV-GBM-1.MethodsKey eligibility criteria for tumor collection were clinical suspicion of newly diagnosed GBM and age 18 to 70 years at the time of surgery. Prior to starting RT/TMZ, key eligibility criteria for intent-to-treat-with-AV-GBM-1 enrollment were: (1) primary GBM confirmed, (2) successful GBM cell culture, (3) collection of sufficient numbers of monocytes (MC) by leukapheresis, (4) Karnofsky Performance Status 70 or greater and (5) plan to treat with concurrent RT/TMZ. AV-GBM-1 was manufactured during RT/TMZ. Interleukin-4 and granulocyte-macrophage colony stimulating factor (GM-CSF) were used to differentiate MC into DC. AV-GBM-1 consists of autologous DC incubated with ATA from the lysate of irradiated GBM cells grown in serum-free media with factors that favor the survival and proliferation of stem cells and early progenitor cells. After recovery from RT/TMZ, over six months patients received up to 8 subcutaneous injections of AV-GBM-1 admixed with 500 μg GM-CSF. The primary objective was to determine if OS was 75% or higher 14.6 months from ITT enrollment, which ended January 2020. The minimum follow-up at the time of analysis was 15.2 months. Secondary endpoints included progression-free survival (PDS) from ITT enrollment and from the first injection.ResultsSuccess rates for cell cultures and sufficient monocyte collections were both 97%. AV-GBM-1 was manufactured for 60/60 (100%). 57 patients received 392 injections; 68% received all 8. The primary adverse events (AE) attributed to AV-GBM-1 were local injection site reactions (16%) and flu-like symptoms (10%). Progression-free survival (PFS) from ITT enrollment is 10.3 months, about 50% longer than reported in four randomized trials with comparable standard therapy arms. PFS from the first injection is 8.3 months, which is 51% and 107% longer than reported in two randomized trials with comparable standard therapy arms. OS was 72% at 12 months, but dropped to 54% at 14.6 months; median OS is 16.0 months.ConclusionsPatent-specific AV-GBM-1 was reliably manufactured and distributed for administration. AV-GBM-1 produced minimal toxicity. PFS was very encouraging but did not translate into OS, perhaps because of discontinuation of treatment after 8 months.Trial Registration[Clinicaltrials.gov NCT03400917]Ethics ApprovalWestern IRB, approval number 20182582Consent n/a
- Published
- 2021
4. 331 Tumor markers associated with increased survival in a phase II trial of dendritic cell/tumor-initiating-cell vaccine AV-GBM-1 in patients with newly diagnosed glioblastoma
- Author
-
Renato V. LaRocca, Frank P.K. Hsu, Robert O. Dillman, Christopher Duma, Candace Hsieh, Santosh Kesari, Gabriel Nistor, Mehrdad Abedi, David Piccioni, Thomas H. Taylor, Jose Carrillo, Robert Aiken, Xiao-Tang Kong, and Daniela A. Bota
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dendritic cell ,Newly diagnosed ,Tumor initiating cell ,medicine.disease ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,business ,RC254-282 ,Glioblastoma - Abstract
BackgroundStandard aggressive therapy of glioblastoma (GBM), which includes maximum safe resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ, is associated with a 25% 2-year overall survival (OS). Adding treatment with AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve OS by inducing and/or enhancing the host anti-GBM immune response. Methylation of the O-6-methylguanine-DNA methyltransferase (MGMT) gene promoter, and mutation of the gene for isocitrate dehydrogenase (IDH) are favorable prognostic markers in newly diagnosed GBM. An objective of a multi-center phase II clinical trial was to determine whether these markers were still prognostic for OS in patients treated with adjunctive AV-GBM-1.MethodsKey eligibility criteria for intent-to-treat (ITT) enrollment were: (1) confirmation of primary GBM, (2) successful GBM cell culture, (3) collection of sufficient numbers of monocytes (MC) by leukapheresis, (4) Karnofsky Performance Status 70 or greater after recovery from surgery, and (5) plan to treat with concurrent RT/TMZ. AV-GBM-1 was manufactured while patients were being treated with RT/TMZ. Interleukin-4 and granulocyte-macrophage colony stimulating factor (GM-CSF) were used to differentiate DC from MC. Each vaccine consisted of autologous DC incubated with ATA from the lysate of irradiated cultured GBM cells grown in serum-free media with factors that favor survival and proliferation of stem cells and early progenitor cells (tumor-initiating cells). After recovery from RT/TMZ, intent was to vaccinate for up to six months with cryopreserved AV-GBM-1 admixed with 500 mg GM-CSF. All patients had testing for MGMT-methylation and IDH-mutation. OS was calculated from date of ITT enrollment.Results60 patients were enrolled during August 2018 to January 2020. MGMT promoter methylation was detected in 21 (35%), mutated IDH in 7 (12%), and one or both in 25 (42%). At a minimum follow-up of 15 months, median OS had not been reached for patients with a methylated MGMT promotor, IDH mutation, or one or both, compared to 14.6 months for 38 with unmethylated MGMT promotor (p=0.026), 14.7 months for 53 with IDH wild-type (p=0.044), and 14.6 months for 35 who had neither (p=0.017). 18-month OS rates were 59% vs 35% for MGMT promotor methylation, 71% vs 40% for IDH mutation and 58% vs 32% for either.ConclusionsBoth MGMT promotor methylation and IDH mutation were associated with a substantial and similar survival benefit in primary GBM patients treated with AV-GBM-1 in addition to standard aggressive therapy.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
5. Insights from immuno-oncology: the Society for Immunotherapy of Cancer Statement on access to IL-6-targeting therapies for COVID-19
- Author
-
Charles G. Drake, Walter J. Urba, David Kaufman, Leisha A. Emens, Jeffrey S. Weber, Alessandra Cesano, Douglas G. McNeel, Ana C. Anderson, Marcela V. Maus, Mario Sznol, Jon M Wiggington, Lisa H. Butterfield, Patrick Hwu, David Parkinson, Ernest C. Borden, James L. Gulley, Michael J. Mastrangelo, Michael B. Atkins, Kim Margolin, Julie R. Brahmer, Thomas F. Gajewski, Robert O. Dillman, Pedro Romero, Daniel S. Chen, Francesco M. Marincola, George J. Weiner, Paolo A. Ascierto, Tanja D. de Gruijl, Michael T. Lotze, Paul M. Sondel, F. Stephen Hodi, Bernard A. Fox, Stefani Spranger, and Howard L. Kaufman
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,viruses ,medicine.medical_treatment ,Immunology ,Betacoronavirus ,Coronavirus Infections/diagnostic imaging ,Coronavirus Infections/drug therapy ,Coronavirus Infections/immunology ,Coronavirus Infections/therapy ,Drug Approval ,Humans ,Immunologic Factors/therapeutic use ,Immunotherapy ,Inflammation/blood ,Inflammation/therapy ,Interleukin-6/agonists ,Neoplasms/therapy ,Pandemics ,Pneumonia, Viral/diagnostic imaging ,Pneumonia, Viral/drug therapy ,Pneumonia, Viral/immunology ,Pneumonia, Viral/therapy ,Receptors, Interleukin-6/agonists ,immunomodulation ,inflammation mediators ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Intensive care ,Pandemic ,medicine ,Immunology and Allergy ,Interleukin 6 ,RC254-282 ,Pneumonitis ,Pharmacology ,biology ,business.industry ,virus diseases ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Hypoxia (medical) ,medicine.disease ,030104 developmental biology ,Editorial ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Respiratory virus ,medicine.symptom ,business - Abstract
The hypoxia and profound inflammatory response associated with the pneumonitis observed with the severe acute respiratory virus coronavirus-2 (SARS-COV-2) virus responsible for the recent COVID-19 pandemic has overwhelmed intensive care facilities in the epicenters of infection including Wuhan
- Published
- 2020
- Full Text
- View/download PDF
6. Randomized phase II trial of autologous dendritic cell vaccines versus autologous tumor cell vaccines in metastatic melanoma: 5-year follow up and additional analyses
- Author
-
Andrew N. Cornforth, Carol DePriest, Gabriel Nistor, Edward F. McClay, Robert O. Dillman, and Thomas Amatruda
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Autologous tumor cell lines ,Immunology ,Metastatic melanoma ,Cancer Vaccines ,Transplantation, Autologous ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Internal medicine ,Clinical endpoint ,Immunology and Allergy ,Medicine ,Humans ,Hypersensitivity, Delayed ,Adverse effect ,Melanoma ,Pharmacology ,Patient-specific vaccines ,business.industry ,Dendritic-cell vaccines ,Dendritic cell ,Dendritic Cells ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Autologous tumor cell ,Survival Analysis ,Tumor-cell vaccines ,Vaccination ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Cytokines ,Female ,business ,Ex vivo ,Research Article - Abstract
Background Despite improved survival following checkpoint inhibitors, there is still a potential role for anti-cancer therapeutic vaccines. Because of biological heterogeneity and neoantigens resulting from each patient’s mutanome, autologous tumor may be the best source of tumor-associated antigens (TAA) for vaccines. Ex vivo loading of autologous dendritic cells with TAA may be associated with superior clinical outcome compared to injecting irradiated autologous tumor cells. We conducted a randomized phase II trial to compare autologous tumor cell vaccines (TCV) and autologous dendritic cell vaccines (DCV) loaded with autologous TAA. Methods Short-term autologous tumor cell lines were established from metastatic tumor. Vaccines were admixed with 500 micrograms of GM-CSF and injected weekly for 3 weeks, then at weeks 8, 12,16, 20, and 24. The primary endpoint was overall survival. Secondary objectives were identification of adverse events, and results of delayed type hypersensitivity (DTH) reactions to intradermal tumor cell injections. Results Forty-two patients were randomized. All were followed from randomization until death or for five years; none were lost to follow-up. DCV was associated with longer survival: median 43.4 versus 20.5 months (95% CI, 18.6 to > 60 versus 9.3 to 32.3 months) and a 70% reduction in the risk of death (hazard ratio = 0.304, p = 0.0053, 95% CI, 0.131 to 0.702). Tumor DTH reactions were neither prognostic nor predictive. The most common treatment-related adverse events were mild to moderate local injection site reactions and flu-like symptoms; but grade 2 treatment-related adverse events were more frequent with TCV. Serum marker analyses at week-0 and week-4 showed that serum markers were similar at baseline in each arm, but differed after vaccination. Conclusions This is the only human clinical trial comparing DCV and TCV as platforms for autologous TAA presentation. DCV was associated with minimal toxicity and long-term survival in patients with metastatic melanoma. DTH to autologous tumor cells was neither prognostic for survival nor predictive of benefit for either vaccine. Trial registration Clinical trials.gov NCT00948480 retrospectively registered 28 July 2009. Electronic supplementary material The online version of this article (10.1186/s40425-018-0330-1) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
7. 332 Tumor collection and establishment of tumor-initiating cell cultures as antigen source for AV-GBM-1 dendritic cell vaccines for a phase II trial in patients with newly diagnosed glioblastoma
- Author
-
Candace Hsieh, Robert O. Dillman, Frank P.K. Hsu, Gabriel Nistor, David Piccioni, Christopher Duma, Santosh Kesari, Jose Carrillo, Robert Aiken, Thomas H. Taylor, Daniela A. Bota, Xiao-Tang Kong, Renato V. LaRocca, and Mehrdad Abedi
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Dendritic cell ,Newly diagnosed ,Tumor initiating cell ,medicine.disease ,Oncology ,Antigen ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,In patient ,business ,Glioblastoma - Abstract
BackgroundDespite standard aggressive therapy (maximum safe surgical resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ), then maintenance TMZ), 2-year survival is only about 25% for patients with newly diagnosed primary glioblastoma (GBM). Adding AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve survival. One objective of a multi-center phase II clinical trial was to determine the feasibility of collecting fresh GBM and establishing short-term cell cultures of GBM tumor-initiating cells (TIC) to serve as ATA source.MethodsKey eligibility criteria for tumor collection were (1) clinical suspicion of new primary GBM, (2) age 18 to 70 years (3) tentative agreement to undergo a leukapheresis procedure after recovery from surgery, and (4) tentative plans for RT/TMZ. Fresh tumor was placed in media and shipped in a transport kit by overnight courier to AIVITA where a cell suspension was placed in culture and incubated in serum-free medium with factors that favor survival and proliferation of TICS (stem cells and early progenitor cells). The intent was to produce a patient-specific DC-ATA vaccine by incubating a lysate of irradiated TICs with autologous DC for subsequent subcutaneous injection.ResultsPatients were enrolled from five sites in California, one in Kentucky and one in New Jersey. Tumors were collected between August 2018 and January 2020. 106 patients consented for tumor collection, but 15 were not GBM, 4 had insufficient tissue to send, 2 patients withdrew consent, 4 were ineligible because of age, and 1 was ineligible because of autoimmune disease. Of the 80 GBM tumors that were placed into culture, 7 were discontinued because of patient withdrawal. 71/73 (97%) resulted in a successful cell culture; two were unsuccessful because of contamination. 60/71 subsequently consented for intent-to-treat ; 46/60 (77%) had cells in culture for 28 days or less, 11 were in culture for 30 to 35 days, and the remaining 3 were cultured 46, 54, and 55 days. The average number of cells per culture at the time of irradiation was 14.0 million (range 0.78 to 63.3 million). 58/60 (97%) yielded more than 1 million TICs for irradiation for the tumor cell lysate; 36/60 (60%) had more than 10 million cells irradiated. 57 patients were subsequently treated with AV-GBM-1 after recovery from RT/TMZ.ConclusionsSelf-renewing GBM TIC cultures can be reliably and rapidly established for use as the antigen source for personal DC-ATA vaccines.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
- Full Text
- View/download PDF
8. 335 Leukaphereses to obtain monocytes to produce dendritic cells in manufacturing of personal autologous AV-GBM-1 vaccines in a phase II trial in patients with newly diagnosed glioblastoma
- Author
-
Gabriel Nistor, Xiang-Tang Kong, Thomas H. Taylor, David Piccione, Robert O. Dillman, Jose Carrillo, Candace Hsieh, Christopher Duma, Daniela A. Bota, Santosh Kesari, Robert Aiken, Renato V. LaRocca, Mehrdad Abedi, and Frank P.K. Hsu
- Subjects
Pharmacology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Newly diagnosed ,Leukapheresis ,Peripheral blood mononuclear cell ,Surgery ,Clinical trial ,Radiation therapy ,Oncology ,Antigen ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 ,medicine.drug - Abstract
BackgroundFor patients with newly diagnosed primary glioblastoma (GBM), maximum safe surgical resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ results in a 2-year survival of only 25%. Adding treatment with AV-GBM-1, a personal vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA), may improve survival. One objective of a multi-center phase II clinical trial was to determine the feasibility of collecting sufficient monocytes (MC) from which to generate DC for pulsing with ATA from GBM tumor-initiating cells (TIC).MethodsPeripheral blood mononuclear cells were collected by leukapheresis per local standard operating procedures, then shipped by overnight courier to the AIVITA laboratory in Irvine, CA. The product was enriched for MC using the Elutra® Cell Separation System (Terumo, Lakewood, CO.). If fewer than 450 million MC were collected, an additional leukapheresis was allowed. MC were cryopreserved in liquid nitrogen and subsequently thawed and incubated in media containing granulocyte-macrophage colony-stimulating factor and interleukin-4 to differentiate MC into DC. Batches of patient-specific AV-GBM-1 were produced by incubating autologous DC with a lysate of irradiated TICs and aliquoted into individual doses.ResultsPatients enrolled from five sites in California, one in Kentucky and one in New Jersey. 65 patients underwent 77 leukapheresis procedures between September 2018 and February 2020; 54 underwent a single pheresis, 10 two phereses, and 1 three (all unsuccessful). The average time from surgical resection to first pheresis was 26 days (range 6 to 90; 64/65 within 51 days). 63/65 (97%) had sufficient MC collected, 53/65 (82%) from a single leukapheresis; 10 required a second procedure. The interval from surgery to first pheresis was the same for those for whom MC collections were satisfactory after one pheresis compared to those who required more than one. The success rate for MC collection for East-coast sites was 14/15 versus 52/62 for West-coast sites (p=0.68); so, longer shipping distance was not an issue. 60 patients who enrolled with intent-to-treat had an average of 1.7 billion monocytes cryopreserved, which were subsequently thawed and differentiated into DC. An average of 750 million DC were incubated with ATA for the final DC-ATA product.ConclusionsLeukapheresis procedures reliably resulted in collection of sufficient numbers of monocytes to generate DC and large batches of personal AV-GBM-1 vaccines. Success after a single leukapheresis was not related to the interval from surgery to pheresis procedure, or distance from the processing site.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
- Full Text
- View/download PDF
9. 333 Changes in proteomic markers after injections of personal AV-GBM-1 dendritic cell/tumor initiating cell vaccines in a phase II trial in patients with newly diagnosed glioblastoma
- Author
-
Thomas H. Taylor, Candace Hsieh, Xiao-Tang Kong, Frank P.K. Hsu, Gabriel Nistor, Christopher Duma, Santosh Kesari, Jose Carrillo, Aleksandra J. Poole, David Piccioni, Daniela A. Bota, Renato V. LaRocca, Mehrdad Abedi, Robert Aiken, and Robert O. Dillman
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Newly diagnosed ,Dendritic cell ,medicine.disease ,Tumor initiating cell ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,business ,RC254-282 ,Glioblastoma - Abstract
BackgroundDespite standard aggressive therapy, including maximum safe surgical resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ, survival is still extremely poor for patients with newly diagnosed primary glioblastoma (GBM). Adding treatment with AV-GBM-1, a personal vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve survival. One objective of a multi-center phase II clinical trial was to determine changes in blood proteomics before and after injections of AV-GBM-1.MethodsAV-GBM-1 consists of autologous DC incubated with ATA from a lysate of irradiated autologous GBM cells that had been placed in culture and incubated in serum-free medium with factors that favor the survival and proliferation of stem cells and early progenitor cells. After recovery from RT/TMZ, GBM patients were injected subcutaneously with AV-GBM-1 admixed in granulocyte-macrophage colony-stimulating factor (GM-CSF) at weeks 1, 2, 3, 8, 12, 16, 20, and 24. Blood samples obtained at baseline (week-0), just prior to the third injection (week-2) and just prior to the fourth injection (week-8), were cryopreserved and subsequently analyzed for 448 proteomic markers using quantitative, multiplex enzyme-linked immunosorbent assays (Raybiotech, Inc., Norcross, GA.). In this preliminary analysis the averages of paired samples for each time point were determined and compared using the student T-Test with a focus on differences of pResultsPatients were enrolled from five sites in California, and one each in Kentucky and New Jersey. 57 patients were treated during November 2018 to October 2020. Paired samples from all three time points were available for 49 patients. After two weekly injections there were increases in thymus-and activation-regulated chemokine (TARC, CCL17), the chemotactic protein chemerin, lipocalin-2, (expressed by macrophages and epithelium in response to inflammation) and angiopoietin-1 (suppressor of vascular inflammation), and decreases in thrombospondin-5 (possibly involved in synaptogenesis in brain repair), angiotensinogen (a precursor of all angiotensin peptides), and beta-fibroblast growth factor (important in tissue repair). The increase in TARC (pConclusionsIf there were humoral changes in proteins associated with Th1 and Th2 responses, these were no longer present after two weekly vaccinations. More sophisticated analyses of this data set, such as principal component analysis, may be needed to understand the effects of AV-GBM-1.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
- Full Text
- View/download PDF
10. Phase I trial of active specific immunotherapy with autologous dendritic cells pulsed with autologous irradiated tumor stem cells in hepatitis B positive patients with hepatocellular carcinoma
- Author
-
Jessica Cannon, Robert O. Dillman, Jia He, Chengwei Chen, William Wei Cao, Michael Bayer, Xiaojin Wang, Craig Fredrickson, Gabriel Nistor, Qingchun Fu, Xiaosong Chen, and Andrew N. Cornforth
- Subjects
Pharmacology ,Hepatitis ,Cancer Research ,medicine.medical_specialty ,Pathology ,Cirrhosis ,Performance status ,business.industry ,Immunology ,Context (language use) ,Hepatitis C ,medicine.disease ,Autologous tumor cell ,Gastroenterology ,digestive system diseases ,Oncology ,Sargramostim ,Hepatocellular carcinoma ,Internal medicine ,Poster Presentation ,medicine ,Molecular Medicine ,Immunology and Allergy ,business ,medicine.drug - Abstract
Background Active or chronic infections may be barriers to successful vaccines, and patients with hepatitis typically are excluded from clinical trials testing therapeutic anticancer vaccines. One concern is vaccines may exacerbate the underlying infection. Hepatocellular carcinoma (HCC) often arises in the context of viral hepatitis B (HBV) or hepatitis C (HBC), with or without cirrhosis. HCC is seldom cured by standard therapy because of undetectable micrometastatic disease that is present at diagnosis. Effective nontoxic adjunctive treatments might improve outcomes for HCC patients who are at high risk for recurrence and death despite surgical resection of their primary hepatoma. Active specific immunotherapy (ASI) with autologous dendritic cells loaded with antigens from autologous tumor stem cell lines has been associated with promising long-term survival in metastatic melanoma, but patients with HBV or HCV were ineligible. Therefore a Phase I safety trial in HCC patients with HBV and/or HIV was warranted. Patients and methods Patients with previously untreated HCC who were candidates for surgical resection, but not curable by resection or liver transplant were enrolled in Shanghai, China. Patients had a solitary lesion >5 cm in diameter, or 3 lesions with at least one > 3 cm, but no regional or distant metastatic disease. Patients had a good performance status, adequate blood counts and organ function. An autologous tumor cell (TC) line was established from the resected tumor. Irradiated autologous TC were incubated with autologous dendritic cells (DC) to create a patient-specific DC-TC product which was cryopreserved. Approximately eight weeks after one course of transarterial chemoembolization therapy (TACE), patients received three weekly subcutaneous injections of DC-TC suspended in 500 micrograms of sargramostim. Patients were monitored for toxicity for 8 weeks from the start of treatment.
- Published
- 2014
- Full Text
- View/download PDF
11. High-dose IL-2 in metastatic melanoma: better survival in patients who also received patient-specific autologous tumor cell vaccine
- Author
-
Robert O. Dillman, Stephanie E. McClure, and Carol DePriest
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,Metastatic melanoma ,business.industry ,Immunology ,Patient specific ,Autologous tumor cell ,Vaccine therapy ,Text mining ,Internal medicine ,Poster Presentation ,Autologous Tumor Cell Vaccine ,medicine ,Molecular Medicine ,Immunology and Allergy ,In patient ,business ,Survival rate - Abstract
Meeting abstracts Treatment with high-dose Interleukin-2 (IL-2) has been associated with long-term survival in small proportion of metastatic melanoma patients. We recently reported a median survival of 15.6 months, and a 20% 5-year survival rate for 150 such patients who were hospitalized for high
- Published
- 2013
- Full Text
- View/download PDF
12. Phase III randomized, double-blinded, placebo-controlled multicenter trial of melapuldencil-t: autologous dendritic cells loaded with irradiated autologous tumor cells (dc-tc) in gm-csf in patients with metastatic melanoma
- Author
-
Andrew N. Cornforth, Robert O. Dillman, Gabriel Nistor, Michael Bayer, and Dynesha Carbonell
- Subjects
Pharmacology ,Cancer Research ,medicine.drug_class ,business.industry ,Melanoma ,Immunology ,medicine.disease ,Placebo ,Monoclonal antibody ,Immune system ,Oncology ,Multicenter trial ,Poster Presentation ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,business ,Autologous dendritic cells ,Autologous tumor - Abstract
Meeting abstracts Genomic analyses have shown that melanoma patients have hundreds to thousands of non-synonymous mutations including unique tumor associated antigens (TAA) that can be recognized by their immune systems. The efficacy of anti-checkpoint monoclonal antibodies provides further proof
- Published
- 2014
13. Superiority of dendritic cell vaccine vs tumor cell vaccine: survival by stratification subsets in MACVAC randomized Phase II trial of patient-specific vaccines utilizing antigens from autologous melanoma tumor cell lines
- Author
-
Carol DePriest, Robert O. Dillman, Denysha Carbonell, David Burtzo, Edward F. McClay, Thomas Amatruda, Andrew N. Cornforth, Clark Haskins, Robert Weber, and George Semeniuk
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,Randomization ,business.industry ,Melanoma ,medicine.medical_treatment ,Immunology ,Immunotherapy ,medicine.disease ,Antigen ,Internal medicine ,Poster Presentation ,medicine ,Molecular Medicine ,Immunology and Allergy ,Stem cell ,Stage (cooking) ,Lost to follow-up ,business ,Adjuvant - Abstract
In a randomized Phase II trial conducted in patients with metastatic melanoma, superior overall survival (p=0.007) was observed for 18 patients treated with vaccines that consisted of autologous dendritic cells loaded with antigens from irradiated autologous melanoma stem cells, (DC-TC, aka eltrapuldencel-T, NBS20 and CBLS20) compared to 24 patients treated with vaccines consisting of autologous irradiated melanoma stem cells (TC) [ClinicalTrials.gov NCT00436930].[1] Both vaccines were admixed with GM-CSF as an adjuvant. Tumor cell lines that served as the source of patient-specific tumor-associated antigens were derived from metastases resected from patients with stage IV or recurrent stage III melanoma. The treatment schedule consisted of weekly subcutaneous injections for 3 weeks, and then monthly for 5 months. The current analysis was undertaken to determine the treatment effects of DC-TC vs TC in each of the subsets defined by the pre-randomization stratifications that were based on whether patients had measurable or non-measurable disease as defined by RECIST, and whether their most advanced stage of disease at the time of randomization had been stage IV or recurrent stage III disease. At the time of this analysis 5 DC-TC and 3 TC patients had been followed for 5 years; 5 patients (3 TC and 2 DC-TC) were alive but followed less than 5 years (minimum 3.5 years); 29 were deceased. No patients were lost to follow up. The survival results are summarized in Table Table1.1. Although the numbers are small, DC-TC immunotherapy was associated with superior survival in each of the four different subsets defined by the stratification variables. Eltrapuldencel-T has moved forward into a pivotal Phase III trial sponsored by Caladrius BioSciences, Inc. Table 1
- Full Text
- View/download PDF
14. Insights from immuno-oncology: the Society for Immunotherapy of Cancer Statement on access to IL-6-targeting therapies for COVID-19
- Author
-
James L Gulley, Julie R Brahmer, Pedro J Romero, Francesco M Marincola, Charles G Drake, Stefani Spranger, Lisa H Butterfield, Douglas G McNeel, Alessandra Cesano, Thomas F Gajewski, Howard L Kaufman, Bernard A Fox, Leisha A Emens, Mario Sznol, Tanja D de Gruijl, Daniel S Chen, Patrick Hwu, Paolo Antonio Ascierto, Walter J Urba, Jon M Wigginton, Robert O Dillman, Michael B Atkins, Kim A Margolin, Paul M Sondel, Michael T Lotze, Ana Carrizosa Anderson, Ernest C Borden, David Kaufman, Michael J Mastrangelo, Marcela V Maus, David R Parkinson, George J Weiner, Jeffrey S Weber, and F Stephen Hodi Jr
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.