Fig 1 - uploaded by Bernard A Fox
Content may be subject to copyright.
Vaccine and autologous tumor delayed-type hypersensitivity (DTH) skin reactions. Grade II vaccine injection site reaction is shown in A , CD3 ϩ 

Vaccine and autologous tumor delayed-type hypersensitivity (DTH) skin reactions. Grade II vaccine injection site reaction is shown in A , CD3 ϩ 

Contexts in source publication

Context 1
... Cs Nordion Gammacell 3000 irradiator; Kanata, Ontario, Canada) to prevent tumor cell proliferation, and cryopreserved in liquid nitrogen. The total process was completed within 36 hours. Irradiated prostate adenocarci- noma PC-3 cells were used as a control in DTH skin tests (Cell Genesys). Suc- cessful vaccine processing required a minimum yield of three vaccines at 5 ϫ 10 6 tumor cells per vaccine. All proce- dures were performed in compliance with regulatory guidelines for gene therapy. Vaccines were administered intrad- ermally every 2 weeks for a total of three to six vaccinations. The vaccine dose was individualized on the basis of yield, and each dose contained 5 ϫ 10 6 to 100 ϫ 10 6 tumor cells. Patients were strati fi ed into three dose ranges for analysis: 5 ϫ 10 6 to 10 ϫ 10 6 cells per vaccination, 10 ϫ 10 6 to 30 ϫ 10 6 cells per vaccination, and 30 ϫ 10 6 to 100 ϫ 10 6 cells per vaccination. Vaccine and DTH cells were thawed and directly in- jected into the extremities. Autologous tumor DTH cells were administered on the same day on the fi rst and fourth vaccinations and at month 9. PC-3 DTH cells were administered with the fourth vaccination only. Immune response of the vaccine and DTH skin reactions was determined by use of the diameter of induration. Punch biopsy specimens were assessed immu- nohistochemically for CD3, CD4, CD8, and CD1a (a dendritic cell marker) with corresponding monoclonal antibodies (Impath Laboratories, Los Angeles, CA). Serum antibodies against autologous lung tumor (when available); allogeneic lung tumor cell lines 157, 441, 520, 596, 1435, 1437, and 2347; the prostate tumor cell line PC-3; and adenovirus before and after vaccination were assayed by immunoblotting (Cell Genesys). For statistical analysis, the primary end points were safety, manufacturing feasibility, and immunologic activity. Secondary end points were tumor response, disease progression, and survival. Adverse events were recorded by use of National Cancer Institute Com- mon Toxicity Criteria. Manufacturing feasibility assessment included analysis of vaccine yields, viability, GM-CSF secretion, and sterility. Tumor staging was performed at baseline and week 12, and response was evaluated with standard Southwest Oncology Group criteria (23). Tumor responses were con fi rmed by repeat imaging studies more than 4 weeks after the initial response. Progression-free and overall survival were calculated by the Kaplan – Meier method from the date of tumor harvest. Univariable and multivariable association analyses between manufacturing, clinical, and immunologic variables were performed with Spearman ’ s correlation coef fi cient, Wilcoxon signed rank test, Fisher ’ s exact test, and Cox propor- tional hazards regression, depending on the nature of the variables analyzed (continuous or categorical). The as- sumptions for using the Cox propor- tional hazards regression test were met. All statistical tests were two-sided. Eighty-three patients underwent tumor harvest (20 in cohort A, 63 in cohort B) and 43 initiated vaccine treatment (10 in cohort A, 33 in cohort B). Patient baseline characteristics are shown in Table 1. All 10 patients in cohort A completed vaccine treatment. The median number of vaccines administered in cohort B was fi ve. The median size of processed solid tumor was 14 g (range ϭ 0.5 – 96 g), and the median volume of pleural fl uid processed was 675 mL (range ϭ 127 – 2600 mL). Among vaccine-treated patients, the median tumor cell dose was 23 ϫ 10 6 cells (range ϭ 5 ϫ 10 6 to 100 ϫ 10 6 cells), with a GM-CSF secretion level (post-thaw) of 104 ng/(24 h) per 10 6 cells (range ϭ 50 – 1871 ng/(24 h) per 10 6 cells) and viability of 62% (range ϭ 11% – 94%). The median number of days from tumor harvest to vaccine release was 31 and that from harvest to initia- tion of vaccine treatment was 49. Vaccines were successfully manufactured in 80% of patients in cohort A and 81% of patients in cohort B. The majority of manufacturing failures (15 of 16 failures) resulted from an insuf fi cient number of tumor cells; two cases of bacterial contamination were noted. The success rate was higher for solid tumors (82%) than for pleural effusions (53%) ( P ϭ .02). GM-CSF secretion varied by 300- fold between vaccine lots. This variability was not associated with any baseline tumor characteristic evaluated, with the exception of higher levels of GM-CSF secreted from pleural effusions than from solid tumors ( P Ͻ .001). Tumor cells had a broad range of viability after thawing that was not consistently associated with any immunologic or clinical end point. The most common vaccine-related adverse events were local vaccine injection site reactions (93%), followed by fatigue (16%), and nausea (12%) and then by pain, arthralgia, and upper respi- ratory infection (each at 5%). All injection site reactions except one were grade 1 or 2 in severity and consisted of local, self-limited erythema, induration, and pruritis (Fig. 1). Two grade 4 (pericar- dial effusion) and six grade 3 (dyspnea, fatigue, injection site reaction, hypoka- lemia, malignant ascites, and pulmonary embolism) possibly related events were reported. There was no association between vaccine dose and the total number of adverse events or grade 3 or 4 adverse events. Immune response to vaccination was measured by vaccine and DTH skin reactions and the induction of tumor- reactive antibodies. After the fi rst injection, 81% of patients developed vaccine site induration, which increased to 90% after repeated vaccination. Vaccine reaction size was positively associated with vaccine GM-CSF secretion ( P ϭ .01) and increased with repeated vaccinations. Analysis of vaccine site biopsy specimens showed dense in fi ltration with CD4 ϩ and CD8 ϩ T cells, CD1a ϩ dendritic cells, and eosinophils (Fig. 1). Cellular immune response to vaccination was monitored by DTH skin reactions to injections of irradiated, autologous tumor cells or control PC-3 prostate cancer cells. Autologous tumor DTH testing was positive ( 5-mm induration) in four (9%) of 43 patients at baseline (all four were negative on repeat testing). After four vaccinations, 10 (34%) of 29 patients tested positive (Fig. 1). No positive DTH reactions (of a total of 10) were seen at doses of fewer than 10 ϫ 10 6 cells compared with 10 (53%) of 19 at higher doses ( P ϭ .04), suggesting a possible dose – response effect. DTH reactions against PC-3 were present in 15 (50%) of 30 patients. Five of 33 patients induced serum antibodies reactive against autologous tumors after vaccination. This analysis was technically limited by insuf fi cient tumor material from most patients and, therefore, was extended to a panel of seven allogeneic lung cancer cell lines. In 32 (78%) of 41 patients, antibody reactivity was induced against at least one lung cancer line, whereas only 13 (32%) of 41 patients induced reactivity against PC-3. Because residual adenoviral proteins are a component of the fi nal vaccine and might serve as an immunologic adjuvant, we measured the impact of vaccination on adenoviral immunity. Most patients (98%) had anti-adenoviral antibodies before vaccination, and 95% had an increased anti-adenoviral antibody titer after vaccination. No statistically signi fi cant differences were noted between the early- and advanced-stage cohorts in any of the immune response end points. Three patients in cohort B achieved durable, complete tumor regressions lasting 6 months, 18 months, and ongo- ing at 22 months. In addition, there was one minor response (30% decrease in a lung nodule) and two mixed responses; seven patients had stable disease (median duration ϭ 7.7 months; range ϭ 4.7 to Ͼ 28 months). Prior chemotherapy for advanced disease had failed for two of the three complete responders, and two had bronchioloalveolar histology (Fig. 2), a relatively uncommon subtype of NSCLC. Complete responses oc- curred at doses of 6.7 ϫ 10 6 to 10 ϫ 10 6 tumor cells per vaccine and at vaccine GM-CSF secretion rates of 44 – 236 ng/ (24 h) per 10 6 cells. Vaccine viability ranged from 19% to 90% among the six patients with any evidence of tumor regression. Immunologic end points were inconsistent; none of the complete responders developed DTH reactions to autologous tumor, but DTH reactions were detected in two of three patients with minor responses. One complete responder showed an in vitro T-cell response to autologous tumor – pulsed dendritic cells after vaccination (data not shown). Antibody responses against autologous tumor were not measured in the complete respond- ers because of a lack of tumor cells. Six recurrences have been observed among the 10 cohort A patients, with a median follow up of 20 months. There were no statistically signi fi cant associations between immunologic end points and tumor response. Survival analysis was performed on cohort B. The median progression-free survival was 4 months (95% con fi dence interval [CI] ϭ 3 to 6 months), and the median overall survival was 9 months (95% CI ϭ 6 to 12 months) among all 63 patients who underwent tumor harvest. The median progression-free survival was 4 months (95% CI ϭ 2 to 6 months), and the median overall survival was 12 months (95% CI ϭ 6 to 19 months) among the 33 treated patients. Survival at 1 year was 39% (95% CI ϭ 34% to 44%) among all patients who underwent tumor harvest and was 44% (95% CI ϭ 37% to 52%) among treated patients. Vaccine-associated GM-CSF secretion was statistically signi fi cantly associated with survival (Fig. 3). Median survival among patients receiving vaccines secreting GM-CSF at a rate of at least 40 ng/24 h per 10 6 cells was 17 months (95% CI ϭ 6 to 23 months) compared with 7 months (95% CI ϭ 4 to 10 months) for those receiving vaccines secreting less GM-CSF ( P ϭ .028). Corresponding 1-year survivals were 56% and 0%, respectively. This GM- CSF cutoff was predetermined from the threshold required for reliable induction ...
Context 2
... per vaccination, and 30 ϫ 10 6 to 100 ϫ 10 6 cells per vaccination. Vaccine and DTH cells were thawed and directly in- jected into the extremities. Autologous tumor DTH cells were administered on the same day on the fi rst and fourth vaccinations and at month 9. PC-3 DTH cells were administered with the fourth vaccination only. Immune response of the vaccine and DTH skin reactions was determined by use of the diameter of induration. Punch biopsy specimens were assessed immu- nohistochemically for CD3, CD4, CD8, and CD1a (a dendritic cell marker) with corresponding monoclonal antibodies (Impath Laboratories, Los Angeles, CA). Serum antibodies against autologous lung tumor (when available); allogeneic lung tumor cell lines 157, 441, 520, 596, 1435, 1437, and 2347; the prostate tumor cell line PC-3; and adenovirus before and after vaccination were assayed by immunoblotting (Cell Genesys). For statistical analysis, the primary end points were safety, manufacturing feasibility, and immunologic activity. Secondary end points were tumor response, disease progression, and survival. Adverse events were recorded by use of National Cancer Institute Com- mon Toxicity Criteria. Manufacturing feasibility assessment included analysis of vaccine yields, viability, GM-CSF secretion, and sterility. Tumor staging was performed at baseline and week 12, and response was evaluated with standard Southwest Oncology Group criteria (23). Tumor responses were con fi rmed by repeat imaging studies more than 4 weeks after the initial response. Progression-free and overall survival were calculated by the Kaplan – Meier method from the date of tumor harvest. Univariable and multivariable association analyses between manufacturing, clinical, and immunologic variables were performed with Spearman ’ s correlation coef fi cient, Wilcoxon signed rank test, Fisher ’ s exact test, and Cox propor- tional hazards regression, depending on the nature of the variables analyzed (continuous or categorical). The as- sumptions for using the Cox propor- tional hazards regression test were met. All statistical tests were two-sided. Eighty-three patients underwent tumor harvest (20 in cohort A, 63 in cohort B) and 43 initiated vaccine treatment (10 in cohort A, 33 in cohort B). Patient baseline characteristics are shown in Table 1. All 10 patients in cohort A completed vaccine treatment. The median number of vaccines administered in cohort B was fi ve. The median size of processed solid tumor was 14 g (range ϭ 0.5 – 96 g), and the median volume of pleural fl uid processed was 675 mL (range ϭ 127 – 2600 mL). Among vaccine-treated patients, the median tumor cell dose was 23 ϫ 10 6 cells (range ϭ 5 ϫ 10 6 to 100 ϫ 10 6 cells), with a GM-CSF secretion level (post-thaw) of 104 ng/(24 h) per 10 6 cells (range ϭ 50 – 1871 ng/(24 h) per 10 6 cells) and viability of 62% (range ϭ 11% – 94%). The median number of days from tumor harvest to vaccine release was 31 and that from harvest to initia- tion of vaccine treatment was 49. Vaccines were successfully manufactured in 80% of patients in cohort A and 81% of patients in cohort B. The majority of manufacturing failures (15 of 16 failures) resulted from an insuf fi cient number of tumor cells; two cases of bacterial contamination were noted. The success rate was higher for solid tumors (82%) than for pleural effusions (53%) ( P ϭ .02). GM-CSF secretion varied by 300- fold between vaccine lots. This variability was not associated with any baseline tumor characteristic evaluated, with the exception of higher levels of GM-CSF secreted from pleural effusions than from solid tumors ( P Ͻ .001). Tumor cells had a broad range of viability after thawing that was not consistently associated with any immunologic or clinical end point. The most common vaccine-related adverse events were local vaccine injection site reactions (93%), followed by fatigue (16%), and nausea (12%) and then by pain, arthralgia, and upper respi- ratory infection (each at 5%). All injection site reactions except one were grade 1 or 2 in severity and consisted of local, self-limited erythema, induration, and pruritis (Fig. 1). Two grade 4 (pericar- dial effusion) and six grade 3 (dyspnea, fatigue, injection site reaction, hypoka- lemia, malignant ascites, and pulmonary embolism) possibly related events were reported. There was no association between vaccine dose and the total number of adverse events or grade 3 or 4 adverse events. Immune response to vaccination was measured by vaccine and DTH skin reactions and the induction of tumor- reactive antibodies. After the fi rst injection, 81% of patients developed vaccine site induration, which increased to 90% after repeated vaccination. Vaccine reaction size was positively associated with vaccine GM-CSF secretion ( P ϭ .01) and increased with repeated vaccinations. Analysis of vaccine site biopsy specimens showed dense in fi ltration with CD4 ϩ and CD8 ϩ T cells, CD1a ϩ dendritic cells, and eosinophils (Fig. 1). Cellular immune response to vaccination was monitored by DTH skin reactions to injections of irradiated, autologous tumor cells or control PC-3 prostate cancer cells. Autologous tumor DTH testing was positive ( 5-mm induration) in four (9%) of 43 patients at baseline (all four were negative on repeat testing). After four vaccinations, 10 (34%) of 29 patients tested positive (Fig. 1). No positive DTH reactions (of a total of 10) were seen at doses of fewer than 10 ϫ 10 6 cells compared with 10 (53%) of 19 at higher doses ( P ϭ .04), suggesting a possible dose – response effect. DTH reactions against PC-3 were present in 15 (50%) of 30 patients. Five of 33 patients induced serum antibodies reactive against autologous tumors after vaccination. This analysis was technically limited by insuf fi cient tumor material from most patients and, therefore, was extended to a panel of seven allogeneic lung cancer cell lines. In 32 (78%) of 41 patients, antibody reactivity was induced against at least one lung cancer line, whereas only 13 (32%) of 41 patients induced reactivity against PC-3. Because residual adenoviral proteins are a component of the fi nal vaccine and might serve as an immunologic adjuvant, we measured the impact of vaccination on adenoviral immunity. Most patients (98%) had anti-adenoviral antibodies before vaccination, and 95% had an increased anti-adenoviral antibody titer after vaccination. No statistically signi fi cant differences were noted between the early- and advanced-stage cohorts in any of the immune response end points. Three patients in cohort B achieved durable, complete tumor regressions lasting 6 months, 18 months, and ongo- ing at 22 months. In addition, there was one minor response (30% decrease in a lung nodule) and two mixed responses; seven patients had stable disease (median duration ϭ 7.7 months; range ϭ 4.7 to Ͼ 28 months). Prior chemotherapy for advanced disease had failed for two of the three complete responders, and two had bronchioloalveolar histology (Fig. 2), a relatively uncommon subtype of NSCLC. Complete responses oc- curred at doses of 6.7 ϫ 10 6 to 10 ϫ 10 6 tumor cells per vaccine and at vaccine GM-CSF secretion rates of 44 – 236 ng/ (24 h) per 10 6 cells. Vaccine viability ranged from 19% to 90% among the six patients with any evidence of tumor regression. Immunologic end points were inconsistent; none of the complete responders developed DTH reactions to autologous tumor, but DTH reactions were detected in two of three patients with minor responses. One complete responder showed an in vitro T-cell response to autologous tumor – pulsed dendritic cells after vaccination (data not shown). Antibody responses against autologous tumor were not measured in the complete respond- ers because of a lack of tumor cells. Six recurrences have been observed among the 10 cohort A patients, with a median follow up of 20 months. There were no statistically signi fi cant associations between immunologic end points and tumor response. Survival analysis was performed on cohort B. The median progression-free survival was 4 months (95% con fi dence interval [CI] ϭ 3 to 6 months), and the median overall survival was 9 months (95% CI ϭ 6 to 12 months) among all 63 patients who underwent tumor harvest. The median progression-free survival was 4 months (95% CI ϭ 2 to 6 months), and the median overall survival was 12 months (95% CI ϭ 6 to 19 months) among the 33 treated patients. Survival at 1 year was 39% (95% CI ϭ 34% to 44%) among all patients who underwent tumor harvest and was 44% (95% CI ϭ 37% to 52%) among treated patients. Vaccine-associated GM-CSF secretion was statistically signi fi cantly associated with survival (Fig. 3). Median survival among patients receiving vaccines secreting GM-CSF at a rate of at least 40 ng/24 h per 10 6 cells was 17 months (95% CI ϭ 6 to 23 months) compared with 7 months (95% CI ϭ 4 to 10 months) for those receiving vaccines secreting less GM-CSF ( P ϭ .028). Corresponding 1-year survivals were 56% and 0%, respectively. This GM- CSF cutoff was predetermined from the threshold required for reliable induction of antitumor immunity in murine models (24). In a multivariable analysis of prognostic factors for overall survival among treated cohort B patients that included manufacturing (dose, GM-CSF secretion, viability, and solid tumor versus pleural effusion), clinical (performance status and prior chemotherapy), and immunologic (vaccine site reaction, tumor DTH reaction, and antibody induction) parameters, only vaccine-associated GM-CSF secretion was statistically signi fi cantly associated with improved survival. Results of this initial prelim- inary investigation will be assessed prospectively in future studies. NSCLC is not considered an im- mune-sensitive malignancy. However, durable tumor regressions in this trial were seen in three of 33 treated patients with metastatic NSCLC. We are not aware of previous reports in which immune therapy ...
Context 3
... was determined by use of the diameter of induration. Punch biopsy specimens were assessed immu- nohistochemically for CD3, CD4, CD8, and CD1a (a dendritic cell marker) with corresponding monoclonal antibodies (Impath Laboratories, Los Angeles, CA). Serum antibodies against autologous lung tumor (when available); allogeneic lung tumor cell lines 157, 441, 520, 596, 1435, 1437, and 2347; the prostate tumor cell line PC-3; and adenovirus before and after vaccination were assayed by immunoblotting (Cell Genesys). For statistical analysis, the primary end points were safety, manufacturing feasibility, and immunologic activity. Secondary end points were tumor response, disease progression, and survival. Adverse events were recorded by use of National Cancer Institute Com- mon Toxicity Criteria. Manufacturing feasibility assessment included analysis of vaccine yields, viability, GM-CSF secretion, and sterility. Tumor staging was performed at baseline and week 12, and response was evaluated with standard Southwest Oncology Group criteria (23). Tumor responses were con fi rmed by repeat imaging studies more than 4 weeks after the initial response. Progression-free and overall survival were calculated by the Kaplan – Meier method from the date of tumor harvest. Univariable and multivariable association analyses between manufacturing, clinical, and immunologic variables were performed with Spearman ’ s correlation coef fi cient, Wilcoxon signed rank test, Fisher ’ s exact test, and Cox propor- tional hazards regression, depending on the nature of the variables analyzed (continuous or categorical). The as- sumptions for using the Cox propor- tional hazards regression test were met. All statistical tests were two-sided. Eighty-three patients underwent tumor harvest (20 in cohort A, 63 in cohort B) and 43 initiated vaccine treatment (10 in cohort A, 33 in cohort B). Patient baseline characteristics are shown in Table 1. All 10 patients in cohort A completed vaccine treatment. The median number of vaccines administered in cohort B was fi ve. The median size of processed solid tumor was 14 g (range ϭ 0.5 – 96 g), and the median volume of pleural fl uid processed was 675 mL (range ϭ 127 – 2600 mL). Among vaccine-treated patients, the median tumor cell dose was 23 ϫ 10 6 cells (range ϭ 5 ϫ 10 6 to 100 ϫ 10 6 cells), with a GM-CSF secretion level (post-thaw) of 104 ng/(24 h) per 10 6 cells (range ϭ 50 – 1871 ng/(24 h) per 10 6 cells) and viability of 62% (range ϭ 11% – 94%). The median number of days from tumor harvest to vaccine release was 31 and that from harvest to initia- tion of vaccine treatment was 49. Vaccines were successfully manufactured in 80% of patients in cohort A and 81% of patients in cohort B. The majority of manufacturing failures (15 of 16 failures) resulted from an insuf fi cient number of tumor cells; two cases of bacterial contamination were noted. The success rate was higher for solid tumors (82%) than for pleural effusions (53%) ( P ϭ .02). GM-CSF secretion varied by 300- fold between vaccine lots. This variability was not associated with any baseline tumor characteristic evaluated, with the exception of higher levels of GM-CSF secreted from pleural effusions than from solid tumors ( P Ͻ .001). Tumor cells had a broad range of viability after thawing that was not consistently associated with any immunologic or clinical end point. The most common vaccine-related adverse events were local vaccine injection site reactions (93%), followed by fatigue (16%), and nausea (12%) and then by pain, arthralgia, and upper respi- ratory infection (each at 5%). All injection site reactions except one were grade 1 or 2 in severity and consisted of local, self-limited erythema, induration, and pruritis (Fig. 1). Two grade 4 (pericar- dial effusion) and six grade 3 (dyspnea, fatigue, injection site reaction, hypoka- lemia, malignant ascites, and pulmonary embolism) possibly related events were reported. There was no association between vaccine dose and the total number of adverse events or grade 3 or 4 adverse events. Immune response to vaccination was measured by vaccine and DTH skin reactions and the induction of tumor- reactive antibodies. After the fi rst injection, 81% of patients developed vaccine site induration, which increased to 90% after repeated vaccination. Vaccine reaction size was positively associated with vaccine GM-CSF secretion ( P ϭ .01) and increased with repeated vaccinations. Analysis of vaccine site biopsy specimens showed dense in fi ltration with CD4 ϩ and CD8 ϩ T cells, CD1a ϩ dendritic cells, and eosinophils (Fig. 1). Cellular immune response to vaccination was monitored by DTH skin reactions to injections of irradiated, autologous tumor cells or control PC-3 prostate cancer cells. Autologous tumor DTH testing was positive ( 5-mm induration) in four (9%) of 43 patients at baseline (all four were negative on repeat testing). After four vaccinations, 10 (34%) of 29 patients tested positive (Fig. 1). No positive DTH reactions (of a total of 10) were seen at doses of fewer than 10 ϫ 10 6 cells compared with 10 (53%) of 19 at higher doses ( P ϭ .04), suggesting a possible dose – response effect. DTH reactions against PC-3 were present in 15 (50%) of 30 patients. Five of 33 patients induced serum antibodies reactive against autologous tumors after vaccination. This analysis was technically limited by insuf fi cient tumor material from most patients and, therefore, was extended to a panel of seven allogeneic lung cancer cell lines. In 32 (78%) of 41 patients, antibody reactivity was induced against at least one lung cancer line, whereas only 13 (32%) of 41 patients induced reactivity against PC-3. Because residual adenoviral proteins are a component of the fi nal vaccine and might serve as an immunologic adjuvant, we measured the impact of vaccination on adenoviral immunity. Most patients (98%) had anti-adenoviral antibodies before vaccination, and 95% had an increased anti-adenoviral antibody titer after vaccination. No statistically signi fi cant differences were noted between the early- and advanced-stage cohorts in any of the immune response end points. Three patients in cohort B achieved durable, complete tumor regressions lasting 6 months, 18 months, and ongo- ing at 22 months. In addition, there was one minor response (30% decrease in a lung nodule) and two mixed responses; seven patients had stable disease (median duration ϭ 7.7 months; range ϭ 4.7 to Ͼ 28 months). Prior chemotherapy for advanced disease had failed for two of the three complete responders, and two had bronchioloalveolar histology (Fig. 2), a relatively uncommon subtype of NSCLC. Complete responses oc- curred at doses of 6.7 ϫ 10 6 to 10 ϫ 10 6 tumor cells per vaccine and at vaccine GM-CSF secretion rates of 44 – 236 ng/ (24 h) per 10 6 cells. Vaccine viability ranged from 19% to 90% among the six patients with any evidence of tumor regression. Immunologic end points were inconsistent; none of the complete responders developed DTH reactions to autologous tumor, but DTH reactions were detected in two of three patients with minor responses. One complete responder showed an in vitro T-cell response to autologous tumor – pulsed dendritic cells after vaccination (data not shown). Antibody responses against autologous tumor were not measured in the complete respond- ers because of a lack of tumor cells. Six recurrences have been observed among the 10 cohort A patients, with a median follow up of 20 months. There were no statistically signi fi cant associations between immunologic end points and tumor response. Survival analysis was performed on cohort B. The median progression-free survival was 4 months (95% con fi dence interval [CI] ϭ 3 to 6 months), and the median overall survival was 9 months (95% CI ϭ 6 to 12 months) among all 63 patients who underwent tumor harvest. The median progression-free survival was 4 months (95% CI ϭ 2 to 6 months), and the median overall survival was 12 months (95% CI ϭ 6 to 19 months) among the 33 treated patients. Survival at 1 year was 39% (95% CI ϭ 34% to 44%) among all patients who underwent tumor harvest and was 44% (95% CI ϭ 37% to 52%) among treated patients. Vaccine-associated GM-CSF secretion was statistically signi fi cantly associated with survival (Fig. 3). Median survival among patients receiving vaccines secreting GM-CSF at a rate of at least 40 ng/24 h per 10 6 cells was 17 months (95% CI ϭ 6 to 23 months) compared with 7 months (95% CI ϭ 4 to 10 months) for those receiving vaccines secreting less GM-CSF ( P ϭ .028). Corresponding 1-year survivals were 56% and 0%, respectively. This GM- CSF cutoff was predetermined from the threshold required for reliable induction of antitumor immunity in murine models (24). In a multivariable analysis of prognostic factors for overall survival among treated cohort B patients that included manufacturing (dose, GM-CSF secretion, viability, and solid tumor versus pleural effusion), clinical (performance status and prior chemotherapy), and immunologic (vaccine site reaction, tumor DTH reaction, and antibody induction) parameters, only vaccine-associated GM-CSF secretion was statistically signi fi cantly associated with improved survival. Results of this initial prelim- inary investigation will be assessed prospectively in future studies. NSCLC is not considered an im- mune-sensitive malignancy. However, durable tumor regressions in this trial were seen in three of 33 treated patients with metastatic NSCLC. We are not aware of previous reports in which immune therapy was the sole therapy as- sociated with complete and durable regression of refractory metastatic NSCLC lesions, particularly those lasting more than 1 year, as observed in two of three complete responders in our study. Interestingly, two of three treated patients with the bronchioloalveolar sub- type of NSCLC achieved a complete response (Fig. 2). This subtype is clini- cally and ...

Citations

... There was no association between GVAX dose and the total number of adverse events or grade 3 and 4 adverse events. 79 A plasmid encoding both GM-CSF and bi-shRNA furin DNA was transfected into harvested tumor cells via electroporation as part of a vaccination termed FANG (Gradalis, Dallas, TX), which provides the afferent arm of the immune system with a full tumor antigen matrix. 47,48 This vaccine is a combination immune therapy that produces intra and extra-cellular adjuvant GM-CSF and simultaneously expresses an innovative RNA interference (RNAi) moiety and a bifunctional short hairpin RNA-furin (bi-shRNA-furin). ...
Article
Granulocyte macrophage-colony stimulating factor (GM-CSF) is a potent immunomodulatory cytokine that is known to facilitate vaccine efficacy by promoting the development and prolongation of both humoral and cellular immunity. In the past years we have generated a novel codon-optimized GM-CSF gene as an adjuvant. The codon-optimized GM-CSF gene significantly increased protein expression levels in all cells tested and helped in generating a strong immune responses against HIV-1 Gag and HPV-associated cancer. Here, we review the literature dealing with the adjuvant activity of GM-CSF both in animal models and clinical trials. We anticipate that the codon-optimized GM-CSF gene offers a practical molecular strategy for potentiating immune responses to tumor cell-based vaccinations as well as other immunotherapeutic strategies.
... GM-CSF has been shown to enhance the recruitment, activation and maturation of dendritic cells [40]. Among the more intriguing clinical studies, a phase I/II multicenter trial in patients with early and advanced stage non-small-cell lung cancer (NSCLC) investigated the safety and efficacy autologous tumor cells genetically modified to secrete GM-CSF [41]. Three of 33 advanced NSCLC patients experienced durable complete responses lasting 6, 18 and at least 22 months. ...
Article
Approximately nine out of ten breast cancer-related deaths are attributable to metastasis. Yet, less than 4% of breast cancer patients are initially diagnosed with metastatic cancer. Therefore, the majority of breast cancer-related deaths are due to recurrence and progression of non-metastatic disease. There is tremendous clinical opportunity for novel adjuvant strategies, such as immunotherapies, that have the potential to prevent progressive recurrences. In particular, autologous tumor cell-based vaccines (ATCVs) can train a patient’s immune system to recognize and eliminate occult disease. ATCVs have several advantages including safety, multivalency and patient specificity. Furthermore, because lumpectomy or mastectomy is indicated for the vast majority of breast cancer patients, resected tumors offer a readily available, patient-specific source of tumor antigen. Disadvantages of ATCVs include poor immunogenicity and production inconsistencies. This review summarizes recent progress in the development of autologous breast tumor vaccines and offers insight for overcoming existing limitations.
... An adenoviral vector for delivery of hGM-CSF gene was safe in NSCLC 89 and a larger trial in NSCLC suggested a correlation of cell dose to survival. 10 Unfortunately, this approach was hampered by feasibility, since genetic transduction of individual tumors required a median of 50 days from harvest to treatment. A scientific advance was the creation of a "bystander" cell line derived from K562, which is universally major histocompatability complex (MHC) negative. ...
Article
We created a vaccine in which irradiated allogeneic lung adenocarcinoma cells are combined with a bystander K562 cell line transfected with hCD40L and hGM-CSF. By recruiting and activating dendritic cells, we hypothesized that the vaccine would induce tumor regression in metastatic lung adenocarcinoma. Intradermal vaccine was given q14 days×3, followed by monthly ×3. Cyclophosphamide (300 mg/m IV) was administered before the first and fourth vaccines to deplete regulatory T cells. All-trans retinoic acid was given (150/mg/m/d) after the first and fourth vaccines to enhance dendritic cell differentiation. Twenty-four participants were accrued at a single institution from October 2006 to June 2008, with a median age 64 years and median of 4 previous lines of systemic therapy. A total of 101 vaccines were administered. Common toxicities were headache (54%) and site reaction (38%). No radiologic responses were observed. Median overall survival was 7.9 months and median progression-free survival was 1.7 months. Of 14 patients evaluable for immunological study, 5 had peptide-induced CD8 T-cell activation after vaccination. Overall, vaccine administration was feasible in an extensively pretreated population of metastatic lung cancer. Despite a suggestion of clinical activity in the subset with immune response, the trial did not meet the primary endpoint of inducing radiologic tumor regression.
... Initial clinical trials in lung cancer used a patient-specific vaccine platform with intradermal vaccination of irradiated autologous tumor cells that were virally engineered to secrete GM-CSF. 34,35 In the first trial of metastatic NSCLC, GM-CSF was transduced into autologous tumor cells with the use of adenoviral vector before irradiation and patient vaccination. 34 A few clinical responses were observed and several lines of evidence suggested a strong immune response. ...
... In a study that included both early and late stage patients, a similar strategy resulted in similarly promising results -several clinical responses were observed with a similar demonstration of immunological outcomes. 35 In an attempt to produce a vaccine with a more consistent rate of GM-CSF production, the next trial employed the use of a vaccine composed of unmodified, but irradiated autologous tumor cells mixed with a GM-CSF-secreting bystander cell line. 36 This approach eliminated the need for viral transduction and potentially allowed for more precise and higher rates of GM-CSF secretion. ...
Article
Full-text available
Both advanced-stage lung cancer and malignant pleural mesothelioma are associated with a poor prognosis. Advances in treatment regimens for both diseases have had only a modest effect on their progressive course. Gene therapy for thoracic malignancies represents a novel therapeutic approach and has been evaluated in several clinical trials. Strategies have included induction of apoptosis, tumor suppressor gene replacement, suicide gene expression, cytokine-based therapy, various vaccination approaches, and adoptive transfer of modified immune cells. This review considers the clinical results, limitations, and future directions of gene therapy trials for thoracic malignancies.
Article
Full-text available
Over a century ago, it was reported that immunization with embryonic/fetal tissue could lead to the rejection of transplanted tumors in animals. Subsequent studies demonstrated that vaccination of embryonic materials in animals induced cellular and humoral immunity against transplantable tumors and carcinogen-induced tumors. Therefore, it has been hypothesized that the shared antigens between tumors and embryonic/fetal tissues (oncofetal antigens) are the key to anti-tumor immune responses in these studies. However, early oncofetal antigen-based cancer vaccines usually utilize xenogeneic or allogeneic embryonic stem cells or tissues, making it difficult to tease apart the anti-tumor immunity elicited by the oncofetal antigens vs. graft-vs.-host responses. Recently, one oncofetal antigen-based cancer vaccine using autologous induced pluripotent stem cells (iPSCs) demonstrated marked prophylactic and therapeutic potential, suggesting critical roles of oncofetal antigens in inducing anti-tumor immunity. In this review, we present an overview of recent studies in the field of oncofetal antigen-based cancer vaccines, including single peptide-based cancer vaccines, embryonic stem cell (ESC)- and iPSC-based whole-cell vaccines, and provide insights on future directions.
Article
Full-text available
The antitumor response after therapeutic vaccination has a limited effect and seems to be related to the presence of T regulatory cells (Treg), which express the immunoregula­tory molecules CTLA4 and Foxp3. The blockage of CTLA4 using antibodies has shown an effective antitumor response conducing to the approval of the human anti-CTLA4 antibody ipilimumab by the US Food and Drug Administration. On the other hand, Foxp3 is crucial for Treg development. For this reason, it is an attractive target for cancer treatment. This study aims to evaluate whether combining therapeutic vaccination with CTLA4 or Foxp3 gene silencing enhances the antitumor response. First, the “in vitro” cell entrance and gene silencing efficacy of two tools, 2′-O-methyl phosphorotioate-modified oligonucleotides (2′-OMe-PS-ASOs) and polypurine reverse Hoogsteen hairpins (PPRHs), were evaluated in EL4 cells and cultured primary lymphocytes. Following B16 tumor transplant, C57BL6 mice were vaccinated with irradiated B16 tumor cells engineered to produce granulocyte-macrophage colony-stimulating factor (GM-CSF) and were intraperitoneally treated with CTLA4 and Foxp3 2′-OMe-PS-ASO before and after vaccination. Tumor growth, mice survival, and CTLA4 and Foxp3 expression in blood cells were measured. The following results were obtained: 1) only 2′-OMe-PS-ASO reached gene silencing efficacy “in vitro”; 2) an improved survival effect was achieved com­bining both therapeutic vaccine and Foxp3 antisense or CTLA4 antisense oligonucleotides (50% and 20%, respectively); 3) The blood CD4+CD25+Foxp3+ (Treg) and CD4+CTLA4+ cell counts were higher in mice that developed tumor on the day of sacrifice. Our data showed that tumor cell vaccine combined with Foxp3 or CTLA4 gene silencing can increase the efficacy of therapeutic antitumor vaccination.
Chapter
In der Onkologie kommt der Entwicklung neuer experimenteller Therapieverfahren eine grosse Bedeutung zu, da trotz zahlreicher Fortschritte durch konventionelle Chemotherapie bei vielen Krebsarten insbesondere im fortgeschrittenen Stadium keine Heilung erreicht werden kann. Eine Vakzinierung mit tumorspezifischen Immunzellen oder genetisch veränderten Tumorzellen wird derzeit in einer Vielzahl klinischer Phase-I- und Phase-II-Studien erprobt. Therapeutische Ansätze auf der Basis monoklonaler Antikörper und sog. »small molecule drugs«, die vor wenigen Jahren noch als experimentell eingestuft wurden, haben inzwischen z. T. eine feste Indikation im klinischen Alltag.
Chapter
Lung cancer is one of the most common causes of cancer death in the world. There are many risk factors for lung cancer including tobacco smoking, chronic lung disease, race and ethnicity, occupational carcinogen exposure, diet and genetic factors. Until now there have been no effective modalities for the early detection of lung cancer. The National Cancer Institute recently released results from its National Lung Screening Trial (NLST) which showed that low dose CT scans compared to chest x-rays can reduce lung cancer mortality by 20 %. For accurate lung cancer staging, multi-modality approaches are used such as positron emission tomography (PET) scan, computed tomography (CT) scan, endobronchial ultrasound (EBUS) biopsy or mediastinoscope biopsy of mediastinal lymph node, pleural tapping procedure, or video-assisted thoracoscopic surgery (VATS). Multi-modality methods have a vital role in lung cancer treatment including surgery, chemotherapy, radiotherapy, targeted therapy and immunotherapy. Anatomical resection and systematic lymphadenectomy is the treatment of choice for early stage lung cancer. Neo-adjuvant chemotherapy is utilized for resectable N2 disease or adjuvant chemotherapy for pathological N2 disease and unresectable advanced disease. Targeted therapies have already been proven as highly effective and less toxic therapies for positive molecular testing in advanced disease.