Abstract
Lessons Learned
This study showed that carefully selected patients with locally advanced and metastatic forms of malignant melanoma and renal cell carcinoma could potentially have long‐term disease control with a tag‐7 gene‐modified tumor cells‐based vaccine.
Randomized clinical trials in patients whose tumors produce low amounts of immunosuppressive factors are needed to confirm this hypothesis in both the adjuvant and metastatic settings.
Background
Immunotherapy may produce long‐lasting effects on survival and toxicity. The magnitude of efficacy may be dependent on immune factors. We analyzed the results of a phase I/II study of a tag‐7 gene‐modified tumor cells‐based vaccine (GMV) in patients with malignant melanoma (MM) or renal cell carcinoma (RCC) with biomarker analysis of immunosuppressive factors (ISFs) production by their tumor cells.
Methods
From 2001 to 2014, 80 patients received GMV: 68 with MM and 12 with RCC. Treatment in the metastatic setting included 61 patients (MM, 51; RCC, 10), and treatment in the adjuvant setting (after complete cytoreduction) included 19 patients (MM, 17; RCC, 2). Twenty‐six patients were stage III (33%), and 54 (67%) were stage IV. The patients’ tumor samples were transferred to culture, transfected with tag‐7 gene, and inactivated by radiation. The produced product was injected subcutaneously every 3 weeks until progression or 2 years of therapy. ISFs were measured in the supernatants of the tumor cell cultures and used as predictive factors.
Results
No major safety issues or grade 5 adverse events (AEs) were seen. One grade 4 and two grade 3 AEs were registered. No AEs were registered in 89.4% of treatment cycles. No delayed AE was found. The 5‐year overall survival (OS) in the intention‐to‐treat population was 25.1%. There were no differences between MM OS and RCC OS (log rank, p = .44). Median OS in the metastatic setting was 0.7 years and in the adjuvant setting was 3.1 years. Classification trees were built on the basis of ISF production (Fig. 1). The median OS was 6.6 years in the favorable prognosis (FP) group (major histocompatibility complex class I polypeptide‐related sequence A [MICA] level ≤582 pg/mL, n = 15) and 4.6 months in the unfavorable (UF) group (MICA level >582 pg/mL, n = 12; p < .0001). No significant differences were found between classification trees based on ISFs (transforming growth factor β1 [TGF‐β1], interleukin‐10 [IL‐10], and vascular endothelial growth factor [VEGF]). In patients with stage III–IV MM with FP, median OS was 2.3 years, with 31% patients alive at 10 years (Fig. 2) in the UF group (0.4 years; log rank, p = 1.94E−5). No FP patients received modern immunotherapy.
Conclusion
GMV showed high results in carefully selected patients with low ISF (TGF‐β1, IL‐10, and VEGF) production. The method should be further investigated in patients with FP.
Discussion
Progress in immunotherapy revealed the possibility of long‐lasting effects of this treatment modality. Previously reported disappointing results were mostly based on the response rate. In the present work, we assessed long‐term survival data and found results for 10‐year survival comparable to those of patients with MM treated with ipilimumab. Despite inferior results for 3‐year OS in the FP group (42%) in comparison with pembrolizumab (45%), nivolumab (52%), and the combination of ipilimumab with nivolumab (58%), distinct mechanism of action, survival results in selected patients, and presence of biomarker for patients selection promote further development of this technology in the contemporary environment.
The presented results demonstrate the importance of creating rigorous criteria for the selection of patients for cellular immunotherapy, taking into account the biological characteristics of malignant tumors. Several aspects of this phenomenon can be used as a source of biomarkers. They can be measured in the blood or locally in the tumor lesion. Owing to the technological features of the method, we can use a new source of biomarkers—the culture of tumor cells that are used for transfection—because no patients could be treated without this step. However, the selection bias is not excluded from our study. On the other hand, ISFs obtained from the primary tumor also has such bias.
Our study showed high negative predictive and prognostic value for vaccine therapy by ISF level production. All patients with increased ISF levels had low OS. It is important to find out when GMV will be futile, rather than to just find patients who will respond to vaccine treatment. There was no absolutely positive predictive and prognostic value. Only a few patients with low ISF levels had good long‐term outcomes. Future trials are essential for the development of this approach.
Figure 1.

Overall survival of vaccine‐based autologous tumor cells modified with the tag‐7 gene, depending on the major histocompatibility complex class I polypeptide‐related sequence A production.
Trial Information
| Disease | Melanoma |
| Disease | Renal cell carcinoma—clear cell |
| Disease | Renal cell carcinoma—not clear cell |
| Stage of Disease/Treatment | Metastatic/advanced |
| Prior Therapy | No designated number of regimens |
| Type of Study | Phase I/II, cohort study |
| Primary Endpoint | Toxicity |
| Primary Endpoint | Overall survival |
| Secondary Endpoint | Overall response rate |
| Secondary Endpoint | Correlative endpoint |
| Additional Details of Endpoints or Study Design | |
| Overall, 80 patients were included across all study cohorts. Common Terminology Criteria for Adverse Events version 3 was used for safety assessment; RECIST 1.1 and immune‐related response criteria systems were used for final efficacy assessment. | |
| The quantitative content of ISFs was measured in the patient's tumor culture supernatants at early passages before transfection. We assay cytotoxic T‐lymphocyte receptor and NK ligand (MICA), TGF‐β1, IL‐10, and VEGF using an enzyme immunoassay in the “sandwich” variant. Results were obtained as factor concentrations and used as continuous variables in the analysis. | |
| Positive outcome by toxicity was considered two or fewer grade 3–4 related AEs in the first 10 patients and less than 10% grade 3–4 AEs in the overall population. | |
| Positive efficacy was considered 30%+ disease control rate or progression‐free survival (PFS) for 6+ months in the metastatic setting or PFS for 12+ months for the adjuvant setting. | |
| In the final biomarker analysis, patients with melanoma were two groups with sufficient effect (SE) and insufficient effect (iSE) for the predictive biomarkers‐based model development. The SE group included those with complete response (CR), partial response (PR), and stable disease (SD) for more than 6 months for the metastatic setting (III and IV inoperable stages) and PFS for more than 12 months for the adjuvant setting (stages III and IV after complete cytoreduction). The group with iSE included those with progression of the disease (PD), SD for less than 6 months in the metastatic setting, and PFS for less than 12 months in the adjuvant setting. | |
| Investigator's Analysis | The primary safety endpoint was met. Showed activity is selected patients by correlative biomarker. |
Drug Information: MM Therapeutic
| Drug 1 | |
| Generic/Working Name | Tag‐7 gene‐modified inactivated tumor cells |
| Company Name | N.N. Petrov National Medical Research Center of Oncology |
| Drug Type | Vaccine |
| Drug Class | Immune therapy |
| Dose | 10,000,000 per flat dose |
| Route | s.c. |
| Schedule of Administration | Patients received GMV once in 3 weeks subcutaneously in three points in the paravertebral region. One dose consisted of 10 million transfected and inactivated tumor cells. No dose reduction was allowed. |
Dose Escalation: MM Therapeutic
| Dose level | Dose of drug: Tag‐7 gene‐modified inactivated tumor cells | No. enrolled | No. evaluable for toxicity |
|---|---|---|---|
| 1 | 10,000,000 | 47 | 47 |
Drug Information: MM Adjuvant
| Drug 1 | |
| Generic/Working Name | Tag‐7 gene‐modified inactivated tumor cells |
| Company Name | N.N. Petrov National Medical Research Center of Oncology |
| Drug Type | Vaccine |
| Drug Class | Immune therapy |
| Dose | 10,000,000 per flat dose |
| Route | s.c. |
| Schedule of Administration | Patients received GMV once in 3 weeks subcutaneously in three points in the paravertebral region. One dose consisted of 10 million transfected and inactivated tumor cells. No dose reduction was allowed. |
Dose Escalation: MM Adjuvant
| Dose level | Dose of drug: Tag‐7 gene‐modified inactivated tumor cells | No. enrolled | No. evaluable for toxicity |
|---|---|---|---|
| 1 | 10,000,000 | 21 | 21 |
Drug Information: RCC Therapeutic
| Drug 1 | |
| Generic/Working Name | Tag‐7 gene‐modified inactivated tumor cells |
| Company Name | N.N. Petrov National Medical Research Center of Oncology |
| Drug Type | Vaccine |
| Drug Class | Immune therapy |
| Dose | 10,000,000 per flat dose |
| Route | s.c. |
Dose Escalation: RCC Therapeutic
| Dose level | Dose of drug: Tag‐7 gene‐modified inactivated tumor cells | No. enrolled | No. evaluable for toxicity |
|---|---|---|---|
| 1 | 10,000,000 | 10 | 9 |
Drug Information: RCC Adjuvant
| Drug 1 | |
| Generic/Working Name | Tag‐7 gene‐modified inactivated tumor cells |
| Company Name | N.N. Petrov National Medical Research Center of Oncology |
| Drug Type | Vaccine |
| Drug Class | Immune therapy |
| Dose | 10,000,000 per flat dose |
| Route | s.c. |
Dose Escalation: RCC Adjuvant
| Dose level | Dose of drug: Tag‐7 gene‐modified inactivated tumor cells | No. enrolled | No. evaluable for toxicity |
|---|---|---|---|
| 1 | 10,000,000 | 3 | 3 |
Drug Information: Biomarker Finding Cohort
| Drug 1 | |
| Generic/Working Name | Tag‐7 gene‐modified inactivated tumor cells |
| Company Name | N.N. Petrov National Medical Research Center of Oncology |
| Drug Type | Vaccine |
| Drug Class | Immune therapy |
| Dose | 10,000,000 per flat dose |
| Route | s.c. |
Dose Escalation: Biomarker Finding Cohort
| Dose level | Dose of drug: Tag‐7 gene‐modified inactivated tumor cells | No. enrolled | No. evaluable for toxicity |
|---|---|---|---|
| 1 | 10,000,000 | 27 | 27 |
Drug Information: MM Favorable
| Drug 1 | |
| Generic/Working Name | Tag‐7 gene‐modified inactivated tumor cells |
| Company Name | N.N. Petrov National Medical Research Center of Oncology |
| Drug Type | Vaccine |
| Drug Class | Immune therapy |
| Dose | 10,000,000 per flat dose |
| Route | s.c. |
Dose Escalation: MM Favorable
| Dose level | Dose of drug: Tag‐7 gene‐modified inactivated tumor cells | No. enrolled | No. evaluable for toxicity |
|---|---|---|---|
| 1 | 10,000,000 | 11 | 12 |
Drug Information: MM Unfavorable
| Drug 1 | |
| Generic/Working Name | Tag‐7 gene‐modified inactivated tumor cells |
| Company Name | N.N. Petrov National Medical Research Center of Oncology |
| Drug Type | Vaccine |
| Drug Class | Immune therapy |
| Dose | 10,000,000 per flat dose |
| Route | s.c. |
| Schedule of Administration | Patients received GMV once in 3 weeks subcutaneously in three points in the paravertebral region. One dose consisted of 10 million transfected and inactivated tumor cells. No dose reduction was allowed. |
Dose Escalation: MM Unfavorable
| Dose level | Dose of drug: Tag‐7 gene‐modified inactivated tumor cells | No. enrolled | No. evaluable for toxicity |
|---|---|---|---|
| 1 | 10,000,000 | 12 |
Patient Characteristics: MM Therapeutic
| Number of Patients, Male | 22 |
| Number of Patients, Female | 25 |
| Stage |
III — 13 IV — 34 |
| Age | Median (range): 47 (23–73) |
| Number of Prior Systemic Therapies | Median (range): 2 (1–10) |
| Performance Status: ECOG |
0 — 5 1 — 27 2 — 15 3 — 0 Unknown — 0 |
| Other | Metastases localization: lung, 21; liver, 12; lymph nodes, 23; skin, 34; bone, 7 |
| Cancer Types or Histologic Subtypes | Cutaneous melanoma, 46; unknown primary melanoma, 1 |
Patient Characteristics: MM Adjuvant
| Number of Patients, Male | 8 |
| Number of Patients, Female | 13 |
| Stage |
III — 12 IV — 9 |
| Age | Median (range): 47 (21–72) |
| Number of Prior Systemic Therapies | Median (range): 2 (1–6) |
| Performance Status: ECOG |
0 — 12 1 — 9 2 — 0 3 — 0 Unknown — 0 |
| Other | Metastases localization: lymph nodes, 6; skin, 6 |
| Cancer Types or Histologic Subtypes | Cutaneous melanoma, 21 |
Patient Characteristics: RCC therapeutic
| Number of Patients, Male | 8 |
| Number of Patients, Female | 1 |
| Stage | IV — 9 |
| Age | Median (range): 49 (25–71) |
| Number of Prior Systemic Therapies | Median (range): 2 (1–5) |
| Performance Status: ECOG |
0 — 1 1 — 5 2 — 3 3 — 0 Unknown — 0 |
| Other | Metastases localization: lung, 9; liver, 2; lymph nodes, 3; skin, 2; bone, 3 |
| Cancer Types or Histologic Subtypes | Renal cell clear cell cancer, 9 |
Patient Characteristics: RCC Adjuvant
| Number of Patients, Male | 0 |
| Number of Patients, Female | 3 |
| Stage | IV — 9 |
| Age | Median (range): 57 (44–65) |
| Number of Prior Systemic Therapies | Median (range): 2 (1–3) |
| Performance Status: ECOG |
0 — 2 1 — 1 2 — 0 3 — 0 Unknown — 0 |
| Other | Metastases localization: lung, 9; liver, 2; lymph nodes, 3; skin, 2; bone, 3 |
| Cancer Types or Histologic Subtypes | Renal cell cancer, 3 |
Patient Characteristics: Biomarker Finding Cohort
| Number of Patients, Male | 12 |
| Number of Patients, Female | 15 |
| Stage |
III — 11 IV — 14 |
| Age | Median (range): 52 (21–73) |
| Number of Prior Systemic Therapies | Median (range): 2 (0–10) |
| Performance Status: ECOG |
0 — 6 1 — 13 2 — 8 3 — 0 Unknown — 0 |
| Other | This cohort was composed of patients from four primary cohorts (MM adjuvant and therapeutic, RCC adjuvant and therapeutic) with known ISF production in cultures. |
| Cancer Types or Histologic Subtypes |
Cutaneous melanoma, 23 Renal cell cancer, 4 |
Patient Characteristics: MM Favorable
| Number of Patients, Male | 3 |
| Number of Patients, Female | 8 |
| Stage |
III — 6 IV — 5 |
| Age | Median (range): 53 (31–67) |
| Number of Prior Systemic Therapies | Median (range): 2 (1–10) |
| Performance Status: ECOG |
0 — 4 1 — 6 2 — 1 3 — 0 Unknown — 0 |
| Other | This cohort was composed of patients from four primary cohorts (MM adjuvant and therapeutic) with low or medium MICA production (MICA <582 pg/mL) |
| Cancer Types or Histologic Subtypes | Cutaneous melanoma, 10; unknown primary melanoma, 1 |
Patient Characteristics: MM Unfavorable
| Number of Patients, Male | 7 |
| Number of Patients, Female | 5 |
| Stage |
III — 5 IV — 7 |
| Age | Median (range): 48.5 (21–73) |
| Number of Prior Systemic Therapies | Median (range): 2 (1–10) |
| Performance Status: ECOG |
0 — 3 1 — 7 2 — 2 3 — 0 Unknown — 0 |
| Other | This cohort was composed of patients from four primary cohorts (MM adjuvant and therapeutic) with high MICA production (MICA >582 pg/ml) |
| Cancer Types or Histologic Subtypes | Cutaneous melanoma, 13 |
Patient Characteristics: Control
| Number of Patients, Male | 20 |
| Number of Patients, Female | 25 |
| Stage |
III — 14 IV — 31 |
| Age | Median (range): 47 (27–72) |
| Number of Prior Systemic Therapies | Median (range): 2 (1–8) |
| Performance Status: ECOG |
0 — 10 1 — 26 2 — 9 3 — 0 Unknown — 0 |
| Other | This cohort was composed of patients from the two primary cohorts (15 from MM adjuvant and 30 from MM therapeutic) with unknown status of MICA production. |
Primary Assessment Method: MM Therapeutic
| Title | Efficacy |
| Number of Patients Screened | 50 |
| Number of Patients Enrolled | 47 |
| Number of Patients Evaluable for Toxicity | 47 |
| Number of Patients Evaluated for Efficacy | 45 |
| Evaluation Method | RECIST 1.1 |
| Response Assessment CR | n = 0 (0%) |
| Response Assessment PR | n =1 (2.1%) |
| Response Assessment SD | n = 5 (10.6%) |
| Response Assessment PD | n = 39 (83%) |
| Response Assessment OTHER | n = 2 (4.3%) |
| (Median) Duration Assessments TTP | 69 days, CI: 48–89 |
| (Median) Duration Assessments OS | 175 days, CI: 38–311 |
| Outcome Notes |
1Y OS — 32% 2Y OS — 22% 3Y OS — 7% 5Y OS — 7% 10Y OS — 7% |
Primary Assessment Method: MM Adjuvant
| Title | Survival |
| Number of Patients Screened | 25 |
| Number of Patients Enrolled | 21 |
| Number of Patients Evaluable for Toxicity | 21 |
| Number of Patients Evaluated for Efficacy | 21 |
| Evaluation Method | RECIST 1.1 |
| Response Assessment CR | Not applicable |
| Response Assessment PR | Not applicable |
| Response Assessment SD | Not applicable |
| Response Assessment PD | Not applicable |
| Response Assessment OTHER | n = 21 (100%) |
| (Median) Duration Assessments TTP | 258 days, CI: 0–847 |
| (Median) Duration Assessments OS | ‐ |
| Outcome Notes |
Median OS was not reached 1Y OS — 73% 2Y OS — 66% 3Y OS — 53% 5Y OS — 53% 10Y OS — 53% |
Secondary Assessment Method: MM Adjuvant
| Efficacy in unfavorable prognosis melanoma |
Primary Assessment Method for Phase I RCC Therapeutic
| Title | Efficacy |
| Number of Patients Screened | 12 |
| Number of Patients Enrolled | 9 |
| Number of Patients Evaluable for Toxicity | 9 |
| Number of Patients Evaluated for Efficacy | 9 |
| Evaluation Method | RECIST 1.1 |
| Response Assessment CR | n = 0 (0%) |
| Response Assessment PR | n = 2 (22.2%) |
| Response Assessment SD | n = 0 (0%) |
| Response Assessment PD | n = 7 (77.8%) |
| Response Assessment OTHER | n = 0 (0%) |
| (Median) Duration Assessments TTP | 114 days, CI: 0–257 |
| (Median) Duration Assessments OS | 631 days, CI: 0–1,504 |
| Outcome Notes |
1Y OS — 50% 2Y OS — 33% 3Y OS — 33% 5Y OS — 33% 10Y OS — 0% |
Primary Assessment Method: RCC Adjuvant
| Title | Efficacy |
| Number of Patients Screened | 3 |
| Number of Patients Enrolled | 3 |
| Number of Patients Evaluable for Toxicity | 3 |
| Number of Patients Evaluated for Efficacy | 3 |
| Evaluation Method | RECIST 1.1 |
| Response Assessment CR | Not applicable |
| Response Assessment PR | Not applicable |
| Response Assessment SD | Not applicable |
| Response Assessment PD | Not applicable |
| Response Assessment OTHER | n = 3 (100%) |
| (Median) Duration Assessments TTP | 365 days, CI: 64–665 |
| (Median) Duration Assessments OS | 1,009 days |
| Outcome Notes |
1Y OS — 50% 2Y OS — 33% 3Y OS — 33% 5Y OS — 33% 10Y OS — 0% |
Primary Assessment Method: Biomarker Finding Cohort
| Title | Efficacy |
| Number of Patients Screened | 80 |
| Number of Patients Enrolled | 27 |
| Number of Patients Evaluable for Toxicity | 27 |
| Number of Patients Evaluated for Efficacy | 27 |
| Evaluation Method | RECIST 1.1 |
| Response Assessment CR | n = 0 (0%) |
| Response Assessment PR | n = 3 (11.1%) |
| Response Assessment SD | n = 7 (25.9%) |
| Response Assessment PD | n = 12 (44.4%) |
| Response Assessment OTHER | n = 5 (18.5%) |
| (Median) Duration Assessments TTP | 170 days, CI: 87–253 |
| (Median) Duration Assessments OS | 455 days, CI: 0–999 |
| Outcome Notes |
1Y OS — 51% 2Y OS — 36% 3Y OS — 18% 5Y OS — 18% 10Y OS — 18% |
Primary Assessment Method: MM Favorable
| Title | Efficacy |
| Number of Patients Evaluable for Toxicity | 11 |
| Number of Patients Evaluated for Efficacy | 11 |
| Evaluation Method | RECIST 1.1 |
| Response Assessment CR | n = 0 (0%) |
| Response Assessment PR | n = 1 (9%) |
| Response Assessment SD | n = 5 (45%) |
| Response Assessment PD | n = 3 (27%) |
| Response Assessment OTHER | n = 2 (18%) |
| (Median) Duration Assessments TTP | 178 days, CI: 50–306 |
| (Median) Duration Assessments OS | 865 days, CI: 763–966 |
| Outcome Notes |
1Y OS — 100% 2Y OS — 67% 3Y OS — 40% 5Y OS — 40% 10Y OS — 40% |
Primary Assessment Method: MM Unfavorable
| Title | Efficacy |
| Number of Patients Evaluable for Toxicity | 12 |
| Number of Patients Evaluated for Efficacy | 12 |
| Evaluation Method | RECIST 1.1 |
| Response Assessment CR | n = 0 (0%) |
| Response Assessment PR | n = 0 (0%) |
| Response Assessment SD | n = 1 |
| Response Assessment PD | n = 9 |
| Response Assessment OTHER | n = 2 |
| (Median) Duration Assessments TTP | 62 days, CI: 38–86 |
| (Median) Duration Assessments OS | 140 days, CI: 120–160 |
| Outcome Notes |
1Y OS — 8% 2Y OS — 8% 3Y OS — 0% |
Primary Assessment Method: Phase I Control
| Title | Efficacy |
| Number of Patients Evaluable for Toxicity | 12 |
| Number of Patients Evaluated for Efficacy | 12 |
| Evaluation Method | RECIST 1.1 |
| Response Assessment CR | n = 0 (0%) |
| Response Assessment PR | n = 0 (0%) |
| Response Assessment SD | n = 1 |
| Response Assessment PD | n = 9 |
| Response Assessment OTHER | n = 2 |
| (Median) Duration Assessments TTP | 62 days, CI: 38–86 |
| (Median) Duration Assessments OS | 140 days, CI: 120–160 |
| Outcome Notes |
1Y OS — 32% 2Y OS — 22% 3Y OS — 7% 5Y OS — 7% 10Y OS — 7% |
Adverse Events: MM Therapeutic
| All Dose Levels, All Cycles | |||||||
|---|---|---|---|---|---|---|---|
| Name | NC/NA | 1 | 2 | 3 | 4 | 5 | All grades |
| Hemoglobin | 100% | 0% | 0% | 0% | 0% | 0% | 0% |
| Pain ‐ arthralgia | 100% | 0% | 0% | 0% | 0% | 0% | 0% |
| Rash: erythema multiforme (e.g., Stevens‐Johnson syndrome, toxic epidermal necrolysis) | 99% | 0% | 1% | 0% | 0% | 0% | 1% |
| Fatigue (asthenia, lethargy, malaise) | 99% | 1% | 0% | 0% | 0% | 0% | 1% |
| Fever (in the absence of neutropenia, where neutropenia is defined as ANC <1.0 × 10e9/L) | 92% | 6% | 2% | 0% | 0% | 0% | 8% |
| Flu‐like syndrome | 99% | 1% | 0% | 0% | 0% | 0% | 1% |
| Injection site reaction/extravasation changes | 100% | 0% | 0% | 0% | 0% | 0% | 0% |
| Blood/bone marrow ‐ leukocytosis | 100% | 0% | 0% | 0% | 0% | 0% | 0% |
| Pain ‐ myalgia | 99% | 1% | 0% | 0% | 0% | 0% | 1% |
| Nausea | 99% | 1% | 0% | 0% | 0% | 0% | 1% |
| Pruritus/itching | 99% | 0% | 0% | 1% | 0% | 0% | 1% |
| Rash/desquamation | 98% | 1% | 0% | 1% | 0% | 0% | 2% |
| Dermatology/skin ‐ vitiligo | 100% | 0% | 0% | 0% | 0% | 0% | 0% |
| Edema: trunk/genital | 100% | 0% | 0% | 0% | 0% | 0% | 0% |
Abbreviations: ANC, absolute neutrophil count; NC/NA, no change from baseline/no adverse event.
Dose‐Limiting Toxicities for Phase I MM Therapeutic
| Dose level | No. enrolled | No. evaluable for toxicity | No. with a dose‐limiting toxicity |
|---|---|---|---|
| 1 | 51 | 51 | 1 |
Adverse Events: MM Adjuvant
| All Cycles | |||||||
|---|---|---|---|---|---|---|---|
| Name | NC/NA | 1 | 2 | 3 | 4 | 5 | All grades |
| Chills | 99% | 1% | 0% | 0% | 0% | 0% | 1% |
| Fatigue | 99% | 1% | 0% | 0% | 0% | 0% | 1% |
| Fever | 99% | 1% | 0% | 0% | 0% | 0% | 1% |
| Hypotension | 99% | 0% | 0% | 1% | 0% | 0% | 1% |
| Pruritus | 99% | 1% | 0% | 0% | 0% | 0% | 1% |
| Rash/desquamation | 99% | 0% | 1% | 0% | 0% | 0% | 1% |
Abbreviation: NC/NA, no change from baseline/no adverse event.
Dose‐Limiting Toxicities: MM Adjuvant
| Dose level | No. enrolled | No. evaluable for toxicity | No. with a dose‐limiting toxicity |
|---|---|---|---|
| 1 | 17 | 17 | 0 |
Adverse Events: RCC Therapeutic
| All Cycles | |||||||
|---|---|---|---|---|---|---|---|
| Name | NC/NA | 1 | 2 | 3 | 4 | 5 | All grades |
| Fever | 98% | 2% | 0% | 0% | 0% | 0% | 2% |
Abbreviation: NC/NA, no change from baseline/no adverse event.
Dose‐Limiting Toxicities for Phase I RCC Therapeutic
| Dose level | No. enrolled | No. evaluable for toxicity | No. with a dose‐limiting toxicity |
|---|---|---|---|
| 1 | 9 | 9 | 0 |
Adverse Events: RCC Adjuvant
| No adverse events in 50 cycles |
Dose‐Limiting Toxicities for Phase I RCC Adjuvant
| Dose level | No. enrolled | No. evaluable for toxicity | No. with a dose‐limiting toxicity |
|---|---|---|---|
| 1 | 3 | 3 | 0 |
Assessment, Analysis, and Discussion
| Completion | Study completed |
| Investigator's Assessment | Primary safety endpoint met. Showed activity is selected patients by correlative biomarker |
Significant progress has been made in the development of new methods of systemic therapy for patients with malignant melanoma (MM) and renal cell carcinoma (RCC) in recent years. Many drugs came to clinical practice recently. At the time of study conduction, there was no possibility for the patients to receive them. So, presented data lack positive effect from effective subsequent therapy.
Checkpoint inhibitors (ipilimumab, pembrolizumab, nivolumab) have proved to be effective in the treatment of advanced and metastatic MM. At the moment, there are no data on the 10‐year overall survival (OS) rate with the use of these drugs, except ipilimumab (10‐year OS rate was 17%). The use of a vaccine based on tumor cells modified with the tag‐7 gene (GMV) in the general population of patients with MM (68 patients) showed a similar result at the same time interval (10‐year OS: 22%). Moreover, in patients with stage III and IV MM in the favorable prognosis (FP) group according to the major histocompatibility complex class I polypeptide‐related sequence A (MICA) concentration with tag‐7 therapy 10‐year OS was 42%, which is significantly higher than with ipilimumab (17%). Of course, the 3‐year OS FP group by MICA (42%) is inferior to the results of pembrolizumab (45%), nivolumab (52%), and the combination of ipilimumab with nivolumab (58%) for the same period. However, according to 5‐year OS, the results were almost equal (42%) compared with pembrolizumab (43%). We postulate that the gene‐modified vaccine has a different mode of action and could be used to induce immune response rather than unblock it. Combinational or sequential strategies could be the most effective in the clinic. Nevertheless, promising survival results in selected patients, and the existence of a biomarker for deciding which patients to treat, will promote further development of this technology in the contemporary environment.
Rosenberg et al. (2004) used World Health Organization (WHO) criteria to describe an objective response rate for vaccines studies. In his work, only 14 (2.6%) out of 440 patients had an objective response—11 partial responses and 3 complete responses. Nowadays, we know that there is a group of patients with progression of the disease (PD) by WHO criteria who have better survival rates on immunotherapy. These results were observed in approximately 30% of patients. In our study, we found 5% of patients with different responses—2 patients with PD by RECIST later had stable disease (SD) by immune‐related response criteria (irRC), and conversely, one patient has SD by RECIST and PD by irRC. Our data represent a similar incidence of this phenomenon. We can propose that the optimal algorithm of efficacy assessment for immunotherapy, including vaccines, should be developed in future trials.
The presented results demonstrate the importance of creating rigorous criteria for the selection of patients for cellular immunotherapy, taking into account the biological characteristics of malignant tumors. These criteria include neutrophil‐to‐lymphocyte ratio, the monocyte‐to‐lymphocyte ratio, the platelet‐to‐lymphocyte ratio, and programmed death‐ligand 1 (PD‐L1) expression, which showed their predictive and prognostic value. However, there are still concerns about the methodological use of leukocyte factors, and no consensus about the standard threshold of PD‐L1 expression for anti‐programmed cell death protein 1 therapy selection has yet been reached.
Immune escape is a well‐known mechanism for resistance to immunotherapy. Several aspects of this phenomenon can be used as a source of biomarkers. They all can be measured in the blood or locally in the tumor lesion. Each source has its advantages and limitations. Owing to the technological features of the method, we can use a new source of biomarkers—the culture of tumor cells that are used for transfection—because no patients could be treated without this step. However, the selection bias is not excluded when using this approach, because not all tumors could be successfully cultured. On the other hand, immunosuppressive factors (ISFs) obtaining from the primary tumor or peripheral blood also has its own bias.
Our study showed high negative predictive and prognostic value for vaccine therapy by ISF level production. All patients with increased ISF levels had low OS. It is equally important to find out when GMV will be futile as it is to just find patients who will respond to vaccine treatment. However, there was no positive predictive and prognostic value. Only a few patients with low ISF levels had excellent long‐term outcomes.
The immunologically tolerant tumor microenvironment has several mechanisms of resistance. High ISF production, such as MICA, transforming growth factor β1, interleukin‐10, and vascular endothelium growth factor, can be one of them. Both local and systemic levels could be involved. We can propose three scenarios of their actions. The first one is antigen presentation impairment at the injection site. In this scenario, high ISF production by vaccine cells prevents effective induction of immune response. The second one is the same mechanisms in tumor lesions that preclude activated immune cells from actions. The third one is systemic immunosuppression caused by these factors. Yet this mechanism is less possible because we do not see major clinical signs of immunosuppression in these patients. Today, we cannot confirm the exact reason for the inefficacy of vaccines. Future biomarker trials will help to understand it. This can also be used for the combination approaches planning.
Our study demonstrated that carefully selected patients with locally advanced and metastatic forms of MM and RCC could potentially have long‐term disease control with GMV. Randomized clinical trials in patients whose tumors produce low amounts of ISF are highly needed to confirm this hypothesis in both the adjuvant and metastatic settings.
Disclosures
The authors indicated no financial relationships.
Figure
Figure 2.

Graphic expression of the quantitative content of immunosuppressive factors in the supernatants of cultures of tumor cells of patients with sufficient and insufficient clinical effect.
Abbreviations: IL‐10, interleukin‐10; MICA, major histocompatibility complex class I polypeptide‐related sequence A; TGF, transforming growth factor; VEGF, vascular endothelium growth factor.
Acknowledgments
We thank Dr. E.V. Harchenko for help in preparing this manuscript.
No part of this article may be reproduced, stored, or transmitted in any form or for any means without the prior permission in writing from the copyright holder. For information on purchasing reprints contact Commercialreprints@wiley.com. For permission information contact permissions@wiley.com.
Footnotes
- ClinicalTrials.gov Identifier: NCT04180774
- Sponsor: N.N. Petrov National Medical Research Center of Oncology
- Principal Investigator: Irina Aleksandrovna Baldueva
- IRB Approved: Yes
References
- 1. Schadendorf D, Hodi FS, Robert C et al. Pooled analysis of long‐term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol 2015,33:1889–1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Dillman RO. An update on the relevance of vaccine research for the treatment of metastatic melanoma. Melanoma Manag 2017;4:203–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Suriano R, Rajoria S, George AL et al. Follow‐up analysis of a randomized phase III immunotherapeutic clinical trial on melanoma. Mol Clin Oncol 2013;1:466–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Dreno B, Thompson JF, Smithers BM et al. MAGE‐A3 immunotherapeutic as adjuvant therapy for patients with resected, MAGE‐A3‐positive, stage III melanoma (DERMA): A double‐blind, randomised, placebo‐controlled, phase 3 trial. Lancet Oncol 2018;19:916–929. [DOI] [PubMed] [Google Scholar]
- 5. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: Moving beyond current vaccines. Nat Med 2004;10:909–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Hodi FS, Hwu WJ, Kefford R et al. evaluation of immune‐related response criteria and RECIST v1.1 in patients with advanced melanoma treated with pembrolizumab. J Clin Oncol 2016;34:1510–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Amato RJ, Hawkins RE, Kaufman HL et al. Vaccination of metastatic renal cancer patients with MVA‐5T4: A randomized, double‐blind, placebo‐controlled phase III study. Clin Cancer Res 2010;16:5539–5547. [DOI] [PubMed] [Google Scholar]
- 8. Rini B, Stenzl A, Zdrojowy R et al. 17LBA Results from an open‐label, randomized, controlled Phase 3 study investigating IMA901 multipeptide cancer vaccine in patients receiving sunitinib as first‐line therapy for advanced/metastatic RCC. Eur J Cancer 2015;51(suppl 3):S718. [Google Scholar]
- 9. Sashchenko LP, Dukhanina EA, Shatalov YV et al. Cytotoxic T lymphocytes carrying a pattern recognition protein Tag7 can detect evasive, HLA‐negative but Hsp70‐exposing tumor cells, thereby ensuring FasL/Fas‐mediated contact killing. Blood 2007;110:1997–2004. [DOI] [PubMed] [Google Scholar]
- 10. Pardoll DM. Paracrine cytokine adjuvants in cancer immunotherapy. Annu Rev Immunol 1995;13:399–415. [DOI] [PubMed] [Google Scholar]
- 11. Moiseyenko VM, Danilov AO, Baldueva IA et al. Phase I/II trial of gene therapy with autologous tumor cells modified with tag7/PGRP‐S gene in patients with disseminated solid tumors: Miscellaneous tumors. Ann Oncol 2005;16:162–168. [DOI] [PubMed] [Google Scholar]
- 12. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell 2011;144:646–674. [DOI] [PubMed] [Google Scholar]
- 13. Pienta KJ, McGregor N, Axelrod R et al. Ecological therapy for cancer: Defining tumors using an ecosystem paradigm suggests new opportunities for novel cancer treatments. Transl Oncol 2008;1:158–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Russo V, Protti MP. Tumor‐derived factors affecting immune cells. Cytokine Growth Factor Rev 2017;36:79–87. [DOI] [PubMed] [Google Scholar]
- 15. Clerici M, Shearer GM, Clerici E. Cytokine dysregulation in invasive cervical carcinoma and other human neoplasias: Time to consider the TH1/TH2 paradigm. J Natl Cancer Inst 1998;90:261–263. [DOI] [PubMed] [Google Scholar]
- 16. Esebanmen GE, Langridge WHR. The role of TGF‐beta signaling in dendritic cell tolerance. Immunol Res 2017, 65:987–994 [DOI] [PubMed] [Google Scholar]
- 17. Kosiewicz MM, Alard P, Liang S et al. Mechanisms of tolerance induced by transforming growth factor‐beta‐treated antigen‐presenting cells: CD8 regulatory T cells inhibit the effector phase of the immune response in primed mice through a mechanism involving Fas ligand. Int Immunol 2004;16:697–706. [DOI] [PubMed] [Google Scholar]
- 18. Seo N, Hayakawa S, Takigawa M et al. Interleukin‐10 expressed at early tumour sites induces subsequent generation of CD4+ T‐regulatory cells and systemic collapse of antitumour immunity. Immunology 2001;103:449–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Prager GW, Poettler M, Unseld M et al. Angiogenesis in cancer: Anti‐VEGF escape mechanisms. Transl Lung Cancer Res 2012;1:14–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Groh V, Wu J, Yee C et al. Tumour‐derived soluble MIC ligands impair expression of NKG2D and T‐cell activation. Nature 2002;419:734–738. [DOI] [PubMed] [Google Scholar]
- 21. Eisenhauer EA, Therasse P, Bogaerts J et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–247. [DOI] [PubMed] [Google Scholar]
- 22. Wolchok JD, Hoos A, O'Day S et al. Guidelines for the evaluation of immune therapy activity in solid tumors: Immune‐related response criteria. Clin Cancer Res 2009;15:7412–7420. [DOI] [PubMed] [Google Scholar]
- 23. Long GV, Schachter J, Ribas A et al. 4‐year survival and outcomes after cessation of pembrolizumab (pembro) after 2‐years in patients (pts) with ipilimumab (ipi)‐naive advanced melanoma in KEYNOTE‐006. J Clin Oncol 2018;36(suppl 15):9503a. [Google Scholar]
- 24. Robert C, Ribas A, Hamid O et al. Three‐year overall survival for patients with advanced melanoma treated with pembrolizumab in KEYNOTE‐001. J Clin Oncol 2016;34(suppl 15):9503a. [Google Scholar]
- 25. Wolchok JD, Chiarion‐Sileni V, Gonzalez R et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2017;377:1345–1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Bilen MA, Martini DJ, Liu Y et al. The prognostic and predictive impact of inflammatory biomarkers in patients who have advanced‐stage cancer treated with immunotherapy. Cancer 2019;125:127–134. [DOI] [PubMed] [Google Scholar]
- 27. Reck M, Rodríguez‐Abreu D, Robinson AG et al. Pembrolizumab versus chemotherapy for PD‐L1‐positive non‐small‐cell lung cancer. N Engl J Med 2016;375:1823–1833. [DOI] [PubMed] [Google Scholar]
- 28. Topalian SL, Taube JM, Anders RA et al. Mechanism‐driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016;16:275–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
