Asset Publisher

mp-105

print Print Back Back

Adoptive Immunotherapy

Policy Number: MP-105

Latest Review Date: October 2023

Category: Medical                                                                

POLICY:

All adoptive immunotherapy techniques intended to enhance autoimmune effects are considered investigational for the indications included, but not limited to, cancers associated with Epstein-Barr virus, Cytomegalovirus-associated cancers, nasopharyngeal cancer, renal cell carcinoma, gastric cancer, colorectal cancer, hepatocellular carcinoma, non-small-cell lung cancer, melanoma, glioblastoma multiforme, medullary thyroid cancer, pancreatic cancer, and cancers treated with autologous peripheral T lymphocytes containing tumor antigen-specific T cell receptors.

Note: CAR-T Therapy is addressed separately in each of the medical policies noted below:

VP-0319 Kymriah™ (tisagenlecleucel), VP-0333 Yescarta™ (axicabtagene ciloleucel), VP-0558 Tecartus™ (brexucabtagene autoleucel), VP-0590 Breyanzi® (lisocabtagene maraleucel), and VP-0598 Abecma® (idecabtagene vicleucel), VP-0663 Carvykti™ (ciltacabtagene autoleucel)

DESCRIPTION OF PROCEDURE OR SERVICE:

The spontaneous regression of certain cancers (e.g., renal cell carcinoma, melanoma) supports the idea that a patient’s immune system can delay tumor progression and, on rare occasions, can eliminate tumors altogether. These observations have led to research into various immunologic therapies designed to stimulate a patient’s own immune system. Adoptive immunotherapy is a method of activating lymphocytes and/or other types of cells for the treatment of cancer and other diseases. Cells are removed from the patient, processed for some period of time, and then infused back into the patient.

Allogeneic cell transplantation following nonmyeloablative conditioning of the recipient (known as reduced-intensity conditioning or RIC) may also be referred to as “adoptive immunotherapy” in the literature. However, RIC cell transplantation relies on a donor-versus-malignancy effect of donor lymphocytes. In contrast, the adoptive immunotherapy techniques described in this policy enhance autoimmune effects primarily. The use of RIC in stem-cell transplantation is discussed for specific cancers in individual policies related to cell transplantation.

Adoptive immunotherapy uses “activated” lymphocytes as a treatment modality. Both nonspecific and specific lymphocyte activation are used therapeutically. Nonspecific, polyclonal proliferation of lymphocytes by cytokines (immune system growth factors), also called autolymphocyte therapy, increases the number of activated lymphocytes.

T Lymphocytes and Killer Cells

Initially, this treatment was performed by harvesting peripheral lymphokine-activated killer (LAK) cells and activating them in vitro with the T-cell growth factor interleukin-2 (IL-2) and other cytokines. More recent techniques yield select populations of cytotoxic T-lymphocytes with specific reactivity to tumor antigens. Peripheral lymphocytes are propagated in vitro with antigen-presenting dendritic cells that have been pulsed with tumor antigens. Alternatively, innate tumor-infiltrating lymphocytes (TIL) from the tumor biopsy are propagated in vitro with IL-2 and anti-CD3 antibody, a T-cell activator. Expansion of TIL for clinical use is labor intensive and requires laboratory expertise. Only a few cancers are infiltrated by T cells in significant numbers; of these, TIL can be expanded in only approximately 50% of cases. These factors limit the widespread applicability of TIL treatment. Cytokine-induced killer (CIK) cells have recently been recognized as a new type of anti-tumor effector cells, which can proliferate rapidly in vitro, with stronger anti-tumor activity and broader spectrum of targeted tumor than other reported anti-tumor effector cells.

Cellular Therapy and Dendritic Cell Infusions

The major research challenge in adoptive immunotherapy is to develop immune cells with anti-tumor reactivity in quantities sufficient for transfer to tumor-bearing patients. In current trials, two methods are studied: adoptive cellular therapy (ACT) and antigen-loaded dendritic cell infusions.

ACT is “the administration of a patient’s own (autologous) or donor (allogeneic) anti-tumor lymphocytes following a lymphodepleting preparative regimen.” Protocols vary, but include these common steps:

1) lymphocyte harvesting (either from peripheral blood or from tumor biopsy)

2) propagation of tumor-specific lymphocytes in vitro using various immune modulators

3) selection of lymphocytes with reactivity to tumor antigens with enzyme-linked immunosorbent assay (ELISA)

4) lymphodepletion of the host with immunosuppressive agents

5) adoptive transfer (i.e., transfusion) of lymphocytes back into the tumor-bearing host

Dendritic cell-based immunotherapy uses autologous dendritic cells (ADC) to activate a lymphocyte-mediated cytotoxic response against specific antigens in vivo. ADCs harvested from the patient are either pulsed with antigen or transfected with a viral vector bearing a common cancer antigen. The activated ADCs are then transfused back into the patient, where they present antigen to effector lymphocytes (CD4+ T cells, CD8+ T cells, and in some cases, B cells). This initiates a cytotoxic response against the antigen and against any cell expressing the antigen. In cancer immunotherapy, ADCs are pulsed with tumor antigens; effector lymphocytes then mount a cytotoxic response against tumor cells expressing these antigens.

In an attempt to further regulate the host immune system, recent protocols use various cytokines (e.g., IL-7 and IL-15 instead of IL-2) to propagate lymphocytes. Protocols also differ in the extent of host lymphodepletion induced prior to transfusing the lymphocytes to the tumor-bearing host.

KEY POINTS:

The most recent literature search was performed through August 11, 2023.

Summary Of Evidence

Cytotoxic T Lymphocytes

For individuals with Epstein-Barr virus-associated cancers who receive cytotoxic T lymphocytes (CTL), the evidence includes 2 small, prospective noncomparative cohort studies. Relevant outcomes are overall survival (OS), disease-specific survival (DSS), quality of life (QOL), and treatment-related mortality and morbidity. The cohort studies have shown a treatment response to infused cytotoxic T lymphocytes directed against cancer-associated viral antigens. To establish efficacy, the following is needed: large, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with Cytomegalovirus-associated cancers who receive cytotoxic T lymphocytes, the evidence includes a single case series. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. In the absence of a randomized controlled trial (RCT) comparing cytotoxic T lymphocytes with standard of care, no conclusions can be made. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

Cytotoxic-Induced Killer Cells

For individuals with nasopharyngeal carcinoma who receive cytotoxic-induced killer (CIK) cells, the evidence includes a single RCT. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. The RCT reported a numerically favorable but statistically insignificant effect on progression-free survival and overall survival. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with renal cell carcinoma who receive CIK cells, the evidence includes multiple RCTs. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. The largest of the RCTs reported statistically significant gains in progression-free survival and overall survival with CIK cell-based immunotherapy compared with interleukin-2 plus interferon-α-2. This body of evidence is limited by the context of the studies (non-U.S.) and choice of a nonstandard comparator. The other 2 RCTs have also reported response rates in favor of CIK therapy with inconsistent effect on survival. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with gastric cancer who receive CIK cells, the evidence includes 2 meta-analyses encompassing non-randomized trials. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. Both meta-analyses reported statistically significant effect on disease-free survival, overall survival, quality of life, and progression free survival in favor of immunotherapy vs no immunotherapy. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with colorectal cancer (CRC) who receive CIK cells, the evidence includes a single RCT and 2 meta-analyses. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. Results of the RCT showed a statistically significant effect on overall survival in favor of immunotherapy vs chemotherapy alone. A meta-analysis that included both gastric cancer and CRC found improvements in OS and PFS in favor of CIK or CIK cell/dendritic cell-cytokine-induced killer compared to chemotherapy alone; another meta-analysis of prospective and randomized studies of CIK or DC-CIK in patients with CRC also showed improvements in survival outcomes compared to non-CIK/DC-CIK treatments. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with hepatocellular carcinoma who receive CIK cells, the evidence includes meta-analyses that include RCTs and quasi-randomized trials. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. Meta-analyses of these trials have reported improved overall survival rates when compared to conventional therapies alone, but they are limited by inclusion of studies from Asia only and heterogeneity in comparators. This body of evidence is limited by the context of the studies (non-U.S.), small sample sizes, heterogeneous treatment groups, and other methodologic weaknesses. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with non-small-cell lung cancer who receive CIK cells, the evidence includes multiple RCTs and a systematic review. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. A single systematic review of RCTs reported some benefits in median time to progression and median survival time. The included body of evidence trials in the systematic review is limited by the context of the studies (non-U.S.), small sample sizes, heterogeneous treatment groups, and other methodologic weaknesses. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

Tumor-Infiltrating Lymphocytes

For individuals with melanoma who receive tumor infiltrating lymphocytes (TILs), the evidence includes a meta-analysis of randomized and non-randomized trials. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. The meta-analysis evaluating TIL with IL-2 in patients with cutaneous melanoma reported an objective response rate (ORR) of 41%. Pooled 1-year overall survival rates ranged from 46.1% to 56.5% depending on the IL-2 dose level. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with EBV-associated nasopharyngeal carcinoma who receive TILs, the evidence includes an RCT evaluating TILs as adjuvant therapy. Relevant outcomes are OS, DSS, QOL, and treatment-related mortality and morbidity. The RCT evaluating TILs as adjuvant therapy following standard chemoradiation in individuals with EBV-associated nasopharyngeal carcinoma found no difference in PFS or other clinical outcomes compared to patients who received standard chemoradiation alone. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

Dendritic Cells

For individuals with glioblastoma multiforme who receive dendritic cells (DC), the evidence includes a systematic review of observational studies. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. Because of the observational and noncomparative nature of the available evidence, it is difficult to draw any meaningful conclusions. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with non-small-cell lung cancer who receive dendritic cells, the evidence includes 2 RCTs and a meta-analysis. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. The RCTs have generally reported some benefits in response rates and/or survival. The results of a meta-analysis of these trials also reported a statistical significant reduction in the hazard of death. Most trials were from Asia and did not use standard of care as the control arm. This body of evidence is limited by the context of the studies (non-U.S.), small sample sizes, heterogeneous treatment groups, and other methodologic weaknesses. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with medullary thyroid cancer (MTC) who receive dendritic cells, the evidence includes one prospective noncomparative study. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. A small prospective noncomparative study in 10 medullary thyroid cancer patients treated with autologous dendritic cells has been published. There are no RCTs comparing dendritic cell-based adoptive immunotherapy with standard of care and, therefore, no conclusions can be made. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with pancreatic cancer who receive dendritic cells, the evidence includes a small prospective noncomparative study. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. The study reported on treatment outcomes for 5 patients with pancreatic cancer. Because of the noncomparative nature of the available evidence and small sample base, it is difficult to draw any meaningful conclusions. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

Genetically Engineered T Cells

Peripheral T Lymphocytes

For individuals with cancers who receive autologous peripheral T lymphocytes containing tumor antigen-specific T-cell receptors (TCRs), the evidence includes multiple small observational studies. Relevant outcomes are overall survival, disease-specific survival, quality of life, and treatment-related mortality and morbidity. Multiple observational studies have examined autologous peripheral T lymphocytes containing tumor antigen-specific T-cell receptors in melanoma, Hodgkin and non-Hodgkin lymphoma, prostate tumors, and neuroblastoma. Because of the noncomparative nature of the available evidence with a small sample size, it is difficult to draw any meaningful conclusion. To establish efficacy, the following is needed: larger, well-conducted, multicentric trials with adequate randomization procedures, blinded assessments, centralized oversight, and the use of an appropriate standard of care as the control arm showing treatment benefit. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

Practice Guidelines and Position Statements

Guidelines or position statements will be considered for inclusion if they were issued by, or jointly by, a US professional society, an international society with US representation, or National Institute for Health and Care Excellence (NICE). Priority will be given to guidelines that are informed by a systematic review, include strength of evidence ratings, and include a description of management of conflict of interest.

National Comprehensive Cancer Network

Current guidelines from the National Comprehensive Cancer Network do not include recommendations for adoptive immunotherapy to treat cancers of the bladder, central nervous system, head and neck, hepatobiliary system, kidney, pancreatic, stomach, thyroid, melanoma, or non-small-cell lung cancer.

U.S. Preventive Services Task Force Recommendations

Not applicable.

KEY WORDS:

Adoptive immunotherapy (AI), lymphokine-activated killer (LAK) cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, autolymphocyte therapy (ALT), leukophoresis, and interleukin-2 (IL-2), cytotoxic T-lymphocytes, genetically engineered T-cells, cytokine-induced killer cells, T-Cell Receptor Therapy, TCR, adoptive cellular therapy, ACT, CIK, antigen-loaded autologous dendritic cells, ADC.

APPROVED BY GOVERNING BODIES:

There are currently no adoptive immunotherapy products within the scope of this review that are U.S. Food and Drug Administration (FDA)-approved. In 2022, the primary analysis of the Netherlands Cancer Institute-sponsored phase 3 M14TIL randomized controlled trial (NCT02278887) was published by Rohaan et al. This study, comparing autologous TIL therapy with ipilimumab in patients with advanced cutaneous melanoma who had received no more than 1 prior line of therapy, met its primary endpoint of prolonged progression-free survival in TIL recipients. The TIL product was prepared at a local facility and, to date, has not been reported to be associated with regulatory application submissions.

In 2022, a pooled analysis of cohorts enrolled in the phase 2 C-144-01 trial (NCT02360579) was published by Chesney et al. In this analysis conducted in patients with advanced non-uveal melanoma who had received a median 3 prior lines of therapy, lifileucel, an autologous CD4+/CD8+ TIL product, demonstrated an overall response rate of 31.4%; with median follow-up of approximately 27 months, median duration of response had not been reached. On the basis of this trial, a Biologics License Application for lifileucel for patients with advanced melanoma was submitted by Iovance Biotherapeutics and accepted by the FDA for priority review, with a Prescription Drug User Fee Act action date of November 25, 2023.

BENEFIT APPLICATION:

Coverage is subject to member’s specific benefits. Group-specific policy will supersede this policy when applicable.

ITS: Home Policy provisions apply

FEP: Special benefit consideration may apply. Refer to member's benefit plan. 

CURRENT CODING: 

CPT codes:

0708T             Intradermal cancer immunotherapy; preparation and initial injection (effective for dates of service 1/1/22 and after)

0709T             ; each additional injection (effective for dates of 1/1/22 and after)

HCPCS codes:

S2107              Adoptive immunotherapy, i.e., development of specific anti-tumor reactivity (e.g., tumor infiltrating lymphocyte therapy) per course of treatment

REFERENCES:

  1. Bachleitner-Hofmann T, Friedl J, Hassler M et al. Pilot trial of autologous dendritic cells loaded with tumor lysate(s) from allogeneic tumor cell lines in patients with metastatic medullary thyroid carcinoma. Oncol Rep 2009; 21(6):1585-92.
  2. Bedrosian I, Mick R, Xu S et al. Intranodal administration of peptide-pulsed mature dendritic cell vaccines results in superior CD8+ T-cell function in melanoma patients. J Clin Oncol 2003; 21(20):3826-35.
  3. Bollard CM, Gottschalk S, Torrano V, et al. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J Clin Oncol. Mar 10 2014; 32 (8):798-808.
  4. Bregy A, Wong TM, Shah AH et al. Active immunotherapy using dendritic cells in the treatment of glioblastoma multiforme. Cancer Treat Rev 2013; 39(8):891-907.
  5. Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. Mar 20 2013; 5 (177):177ra138.
  6. Cai XR, Li X, Lin JX, et al. Autologous transplantation of cytokine-induced killer cells as an adjuvant therapy for hepatocellular carcinoma in Asia: an update meta-analysis and systematic review. Oncotarget. May 09 2017; 8(19):31318-31328.
  7. Cao J, Kong FH, Liu X, et al. Immunotherapy with dendritic cells and cytokine-induced killer cells for hepatocellular carcinoma: A meta-analysis. World J Gastroenterol. Jul 21 2019; 25(27): 3649-3663.
  8. Chen R, Deng X, Wu H, et al. Combined immunotherapy with dendritic cells and cytokine-induced killer cells for malignant tumors: a systematic review and meta-analysis. Int Immunopharmacol. Oct 2014; 22 (2):451-464.
  9. Chia WK, Teo M, Wang WW, et al. Adoptive T-cell transfer and chemotherapy in the first-line treatment of metastatic and/or locally recurrent nasopharyngeal carcinoma. Mol Ther. Jan 2014; 22 (1):132-139.
  10. Chesney J, Lewis KD, Kluger H, et al. Efficacy and safety of lifileucel, a one-time autologous tumor-infiltrating lymphocyte(TIL) cell therapy, in patients with advanced melanoma after progression on immune checkpoint inhibitors and targeted therapies: pooled analysis of consecutive cohorts of the C-144-01 study. J Immunother Cancer. Dec 2022; 10(12).
  11. Chung MJ, Park JY, Bang S, et al. Phase II clinical trial of ex vivo-expanded cytokine-induced killer cells therapy in advanced pancreatic cancer. Cancer Immunol Immunother. Sep 2014; 63 (9):939-946.
  12. Cui J, Wang N, Zhao H, et al. Combination of radiofrequency ablation and sequential cellular immunotherapy improves progression-free survival for patients with hepatocellular carcinoma. Int J Cancer. Jan 15 2014; 134 (2):342-351.
  13. Dafni U, Michielin O, Lluesma SM, et al. Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: a systematic review and meta-analysis. Ann Oncol. Dec 01 2019; 30(12): 1902-1913.
  14. Du H, Yang J. Zhang Y. Cytokine-induced killer cell/dendritic cell combine with cytokine-induced killer cell immunotherapy for treating advanced gastrointestinal cancer. BMC Cancer. Apr 28 2020; 20(1): 357.
  15. Hirooka Y, Itoh A, Kawashima H et al. A combination therapy of gemcitabine with immunotherapy for patients with inoperable locally advanced pancreatic cancer. Pancreas 2009; 38(3): e69-74.
  16. Hontscha C, Borck Y, Zhou H et al. Clinical trials on CIK cells: first report of the international registry on CIK cells (IRCC). J Cancer Res Clin Oncol 2011; 137(2):305-10.
  17. Howard JM, Nandy K, Woldu SL, et al. Demographic Factors Associated with Non-Guideline-Based Treatment of Kidney Cancer in the United Stated. JAM Netw Open. Jun 01 2021; 4(6): e2112813.
  18. Humphries C. Adoptive cell therapy: Honing that killer instinct. Nature. Dec 19 2013; 504 (7480): S13-15.
  19. Johnson LA, Morgan RA, Dudley ME et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 2009; 114(3):535-46.
  20. Khammari A, Labarriére N, Vignard V et al. Treatment of metastatic melanoma with autologous Melan-A/Mart-1-specific cytotoxic T lymphocyte clones. J Invest Dermatol 2009; 129(12):2835-42.
  21. Kimura H, Iizasa T, Ishikawa A, Shingyouji M, et al. Prospective phase II study of post-surgical adjuvant chemo-immunotherapy using autologous dendritic cells and activated killer cells from tissue culture of tumor-draining lymph nodes in primary lung cancer patients. Anticancer Res, Mar-Apr 2008; 28(2B): 1229-1238.
  22. Lacy MQ, Wettstein P, Gastineau DA et al. Dendritic cell-based idiotype vaccination in post-transplant multiple myeloma. Blood 1999; 94(10 suppl part 1):122a.
  23. Lee JH, Lee JH, Lim YS, et al. Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology. Jun 2015; 148(7):1383-1391 e1386.
  24. Leplina OY, Stupak, VV, Kozlov, YP, et al. Use of interferon-alpha-induced dendritic cells in the therapy of patients with malignant brain gliomas. Bull Exp Biol Med, April 2007; 143(4): 528-534.
  25. Li CMY, Tomita Y, Dhakal B, et al. Use of cytokine-induced killer cell therapy in patients with colorectal cancer: asystematic review and meta-analysis. J Immunother Cancer. Apr 2023; 11(4).
  26. Li JJ, Gu MF, Pan K et al. Autologous cytokine-induced killer cell transfusion in combination with gemcitabine plus cisplatin regimen chemotherapy for metastatic nasopharyngeal carcinoma. J Immunother 2012; 35(2):189-95.
  27. Liang, YJ, Chen QY, Xu JX, et al. A phase II randomised controlled trial of adjuvant tumour-infiltrating lymphocytes forpretreatment Epstein-Barr virus DNA-selected high-risk nasopharyngeal carcinoma patients. Eur J Cancer. Sep 2023;191: 112965.
  28. Liau LM, Ashkan K, Tran DD, et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J Transl Med. May 29 2018; 16 (1):142.
  29. Liu L, Zhang W, Qi X et al. Randomized study of autologous cytokine-induced killer cell immunotherapy in metastatic renal carcinoma. Clin Cancer Res 2012; 18(6):1751-9.
  30. Lovance Biotherapeutics, Inc. Iovance Biotherapeutics Announces U.S. Food and Drug Administration Acceptance of theBiologics License Application of Lifileucel for the Treatment of Advanced Melanoma. May 26, 2023.https://ir.iovance.com/news-releases/news-release-details/iovance-biotherapeutics-announces-us-food-and-drug.
  31. Maude SL, Teachey DT, Porter DL, et al. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood. Jun 25 2015; 125(26):4017-4023.
  32. Motta MR, Castellani S, Rizzi S et al. Generation of dendritic cells from CD14+ monocytes positively selected by immunomagnetic adsorption for multiple myeloma patients enrolled in a clinical trial of anti-idiotype vaccination. Br J Haematol 2003; 121(2):240-50.
  33. National Cancer Institute. SEER Cancer Stat Facts: Kidney and Renal Pelvis Cancer. 2023. https://seer.cancer.gov/statfacts/html/kidrp.html.
  34. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology: bladder cancer. Version 3.2023. Updated May 25, 2023. http://www.nccn.org/professionals/physician_gls/pdf/bladder.pdf
  35. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: central nervous system cancers, Version 1.2023. Updated March 24, 2023. www.nccn.org/professionals/physician_gls/pdf/cns.pdf.
  36. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: biliary tract cancers. Version 2.2023. Updated May 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/btc.pdf.
  37. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: head and neck cancers, Version 2.2023. Updated May 15, 2023.  www.nccn.org/professionals/physician_gls/pdf/head-and-neck.pdf.
  38. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: hepatocellular carcinoma. Version 1.2023. Updated March 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/hcc.pdf.
  39. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: gastric cancer. Version1.2023. Updated March 10, 2023. http://www.nccn.org/professionals/physician_gls/pdf/gastric.pdf.
  40. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: cutaneous melanoma, Version 2.2023. Updated March 10, 2023. www.nccn.org/professionals/physician_gls/pdf/cutaneous_melanoma.pdf.
  41. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: non-small cell lung cancer, Version 3.2023.  Updated April 13, 2023. www.nccn.org/professionals/physician_gls/pdf/nscl.pdf.
  42. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: pancreatic adenocarcinoma, Version 2.2023. Updated June 19, 2023. www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf.
  43. National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: thyroid carcinoma, Version 3.2023. Updated July 27, 2023.  www.nccn.org/professionals/physician_gls/pdf/thyroid.pdf.
  44. Ngo MC, Rooney CM, Howard JM, et al. Ex vivo gene transfer for improved adoptive immunotherapy of cancer. Hum Mol Genet 2011; 20(R1): R93-9.
  45. Ochi T, Fujiwara H, Yasukawa M. Requisite considerations for successful adoptive immunotherapy with engineered T-lymphocytes using tumor antigen-specific T-cell receptor gene transfer. Expert Opin Biol Ther 2011; 11(6):699-713.
  46. Oelke M, Krueger C, and Schneck JP. Technological advances in adoptive immunotherapy.  Drugs of Today 2005; 41(1): 13-21.
  47. Ohtani T, Yamada Y, Furuhashi A, et al. Activated cytotoxic T-lymphocyte immunotherapy is effective for advanced oral and maxillofacial cancers. Int J Oncol. Nov 2014; 45 (5):2051-2057.
  48. Olson DJ, Odunsi K. Adoptive Cell Therapy for Nonhematologic Solid Tumors. J Clin Oncol. Jun 20 2023; 41(18): 3397-3407.
  49. Pinthus JH, Waks T, Malina V et al. Adoptive immunotherapy of prostate cancer bone lesions using redirected effector lymphocytes. J Clin Invest 2004; 114(12):1774-81.
  50. Pule MA, Savoldo B, Myers GD, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med 2008; 14(11):1264-70.
  51. Rohaan MW, Borch TH, van den Berg JH, et al.
  52. Tumor-Infiltrating Lymphocyte Therapy or Ipilimumab in AdvancedMelanoma. N Engl J Med. Dec 08 2022; 387(23): 2113-2125.
  53. Rosenberg SA, RestifoNP, Yang JC et al. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 2008; 8(4):299-308.
  54. Rosenberg SA, Yang JC, Sherry RM et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011; 17(13):4550-7.
  55. Santin AD, Bellone S, Palmieri M et al. Induction of tumor-specific cytotoxicity in tumor infiltrating lymphocytes by HPV16 and HPV18 E7-pulsed autologous dendritic cells in patients with cancer of the uterine cervix. Gynecol Oncol 2003; 89(2):271-80.
  56. Savoldo B, Rooney CM, Di Stasi A et al. Epstein Barr virus specific cytotoxic T lymphocytes expressing the anti-CD30zeta artificial chimeric T-cell receptor for immunotherapy of Hodgkin disease. Blood 2007; 110(7):2620-30.
  57. Schuessler A, Smith C, Beagley L, et al. Autologous T-cell therapy for cytomegalovirus as a consolidative treatment for recurrent glioblastoma. Cancer Res. Jul 1 2014; 74(13):3466-3476.
  58. Shi L, Zhou Q, Wu J et al. Efficacy of adjuvant immunotherapy with cytokine-induced killer cells in patients with locally advanced gastric cancer. Cancer Immunol Immunother 2012; 61(12):2251-9.
  59. Shi SB, Ma TH, Li CH et al. Effect of maintenance therapy with dendritic cells: cytokine-induced killer cells in patients with advanced non-small cell lung cancer. Tumori 2012; 98(3):314-9.
  60. Straten P, Becker JC. Adoptive cell transfer in the treatment of metastatic melanoma. J Invest Dermatol 2009; 129(12):2743-5.
  61. Su Z, Dannull J, Heiser A et al. Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells. Cancer Res 2003; 63(9):2127-33.
  62. Tallen G, Ratei R, Mann G, et al. Long-term outcome in children with relapsed acute lymphoblastic leukemia after time-point and site-of-relapse stratification and intensified short-course multidrug chemotherapy: results of trial ALL-REZ BFM 90. J Clin Oncol. May 10 2010; 28(14):2339-2347.
  63. Tang X, Liu T, Zang X et al. Adoptive cellular immunotherapy in metastatic renal cell carcinoma: a systematic review and meta-analysis. PLoS One 2013; 8(5):e62847.
  64. Tanyi JL, Chu CS. Dendritic cell-based tumor vaccinations in epithelial ovarian cancer: a systematic review. Immunotherapy 2012; 4(10):995-1009.
  65. Till BG, Jensen MC, Wang J et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantel cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 2008; 112(6):2261-71.
  66. Timmerman JM, Czerwinski DK, Davis TA et al. Idiotype-pulsed dendritic cell vaccination for B-cell lymphoma: clinical and immune response in 35 patients. Blood 2002; 99(5):1517-29.
  67. Triozzi PL, Khurram R, Aldrich WA et al. Intratumoral injection of dendritic cells derived in vitro in patients with metastatic cancer. Cancer 2000; 89(12):2646-54.
  68. Wang M, Cao JX, Pan JH, et al. Adoptive immunotherapy of cytokine-induced killer cell therapy in the treatment of non-small cell lung cancer. PLoS One. 2014; 9(11):e112662.
  69. Wang X, Tang S, Cui X, et al. Cytokine-induced killer cell/dendritic cell-cytokine-induced killer cell immunotherapy for the postoperative treatment of gastric cancer: A systematic review and meta-analysis. Medicine (Baltimore). Sep 2018;97(36):e12230.
  70. Xie F, Zhang X, Li H et al. Adoptive immunotherapy in postoperative hepatocellular carcinoma: a systemic review. PloS One 2012; 7(8):e42879.
  71. Yang L, Ren B, Li H et al. Enhanced antitumor effects of DC-activated CIKs to chemotherapy treatment in a single cohort of advanced non-small-cell lung cancer patients. Cancer Immunol Immunother 2013; 62(1):65-73.
  72. Yarza R, Bover M, Herrera-Juarez M, et al. Efficacy of T-Cell Receptor-Based Adoptive Cell Therapy in Cutaneous Melanoma: A Meta-Analysis. Oncologist. Jun 02 2023; 28(6): e406-e415.
  73. Zhang Y, Wang J, Wang Y, et al. Autologous CIK cell immunotherapy in patients with renal cell carcinoma after radical nephrectomy. Clin Dev Immunol. 2013; 2013: 195691.
  74. Zhang G, Zhao H, Wu J, et al. Adoptive immunotherapy for non-small cell lung cancer by NK and cytotoxic T lymphocytes mixed effector cells: retrospective clinical observation. Int Immunopharmacol. Aug 2014; 21 (2):396-405.
  75. Zhao H, Wang Y, Yu J, et al. Autologous cytokine-induced killer cells improves overall survival of metastatic colorectal cancer patients: results from a phase II clinical trial. Clin Colorectal Cancer. Sep 2016; 15(3):228-235.
  76. Zhao X, Zhang Z, Li H, et al. Cytokine induced killer cell-based immunotherapies in patients with different stages of renal cell carcinoma. Cancer Lett. Jul 1 2015; 362(2):192-198.
  77. Zhong JH, Ma L, Wu LC et al. Adoptive immunotherapy for postoperative hepatocellular carcinoma: a systematic review. Int J Clin Pract 2012; 66(1):21-7.

POLICY HISTORY:

Medical Policy Group, April 2003 (1)

Medical Policy Administration Committee, August 2003

Available for comment August 11-September 25, 2003

Medical Policy Group, April 2005 (1)

Medical Policy Group, April 2007 (1)

Medical Policy Group, January 2009 (1)

Medical Policy Group, June 2011 (1): Update to Key Points and References

Medical Policy Group, March, 2012 (1): Update to Key Points and References related to MPP update; no change in policy statement

Medical Policy Group, September 2013 (1): Update to the policy statement under adoptive cellular therapy was changed to include cytokine-induced killer (CIK) cells; however, the intent of both policy statement (i.e., investigational) is unchanged; update to Description, Key Points and References

Medical Policy Administration Committee, September 2013

Medical Policy Panel, December 2013

Medical Policy Group, January 2014 (1): Update to Key Points and References; no change in policy statement

Medical Policy Panel, December 2014

Medical Policy Group, December 2014 (4): Updates to Description, Key Points, Approved Governing Bodies, Key Words and References.  Update to policy statement adding “cytotoxic T-lymphocytes”, “autologous” dendritic cells and “genetically engineered T-cells”. Also updated Policy section to include other applications of adoptive immunotherapy are considered investigational

Medical Policy Panel, December 2015

Medical Policy Group, January 2016 (3): Updates to Description, Key Points & References; no change in policy statement.

Medical Policy Group, March 2016 (3): Update to Key Words. No change in policy statement.

Medical Policy Group, August 2017 (2): Updates to Key Words, Approved by Governing Bodies, Current Coding, and References; no change in policy statement.

Medical Policy Group, October 2017 (2): Updates to Key Words, Approved by Governing Bodies, Current Coding, and References; no change in policy section.

Medical Policy Group, November 2017 (2): Updates to Key Points and References; No change in policy statement.

Medical Policy Group, December 2017: Annual Coding Update 2018.  Added new HCPCS code Q2040 to the Current Coding section.

Medical Policy Group, December 2017 (2): Removed all CAR-T related content and made a note to refer to medical policies #675 Kymriah and #676 Yescarta.

Medical Policy Panel, July 2018

Medical Policy Group, August 2018 (2): Updates to Key Points and References; no change in policy statement.

Medical Policy Group, March 2019 (2): Updates to Key Points and References; no change in policy statement.

Medical Policy Panel, October 2019

Medical Policy Group, November 2019 (2): Updates to Key Points; no change in policy statement.

Medical Policy Panel, October 2020

Medical Policy Group, October 2020 (2): Updates to Key Points and Reference; no change in Policy Statement; note added for allogeneic cell transplantation is not adoptive immunotherapy.

Medical Policy Panel, October 2021

Medical Policy Group, October 2021 (2): Updates to Key Points and References; Policy statement updated to remove “not medically necessary,” no change to intent.

Medical Policy Group, December 2021 (2): 2022 Annual Coding Update, new codes 0708T and 0709T added to the Current Coding section.           

Medical Policy Panel, October 2022

Medical Policy Group, October 2022 (2): Updates to Key Points and References; no change in policy statement.

Medical Policy Panel, October 2023

Medical Policy Group, October 2023 (2): Updates to Key Points, Approved by Governing Bodies, Benefit Application, and References; no change in Policy statement. 

This medical policy is not an authorization, certification, explanation of benefits, or a contract. Eligibility and benefits are determined on a case-by-case basis according to the terms of the member’s plan in effect as of the date services are rendered. All medical policies are based on (i) research of current medical literature and (ii) review of common medical practices in the treatment and diagnosis of disease as of the date hereof. Physicians and other providers are solely responsible for all aspects of medical care and treatment, including the type, quality, and levels of care and treatment.

This policy is intended to be used for adjudication of claims (including pre-admission certification, pre-determinations, and pre-procedure review) in Blue Cross and Blue Shield’s administration of plan contracts.

The plan does not approve or deny procedures, services, testing, or equipment for our members. Our decisions concern coverage only. The decision of whether or not to have a certain test, treatment or procedure is one made between the physician and his/her patient. The plan administers benefits based on the member’s contract and corporate medical policies. Physicians should always exercise their best medical judgment in providing the care they feel is most appropriate for their patients. Needed care should not be delayed or refused because of a coverage determination.

As a general rule, benefits are payable under health plans only in cases of medical necessity and only if services or supplies are not investigational, provided the customer group contracts have such coverage.

The following Association Technology Evaluation Criteria must be met for a service/supply to be considered for coverage:

1. The technology must have final approval from the appropriate government regulatory bodies;

2. The scientific evidence must permit conclusions concerning the effect of the technology on health outcomes;

3. The technology must improve the net health outcome;

4. The technology must be as beneficial as any established alternatives;

5. The improvement must be attainable outside the investigational setting.

Medical Necessity means that health care services (e.g., procedures, treatments, supplies, devices, equipment, facilities or drugs) that a physician, exercising prudent clinical judgment, would provide to a patient for the purpose of preventing, evaluating, diagnosing or treating an illness, injury or disease or its symptoms, and that are:

1. In accordance with generally accepted standards of medical practice; and

2. Clinically appropriate in terms of type, frequency, extent, site and duration and considered effective for the patient’s illness, injury or disease; and

3. Not primarily for the convenience of the patient, physician or other health care provider; and

4. Not more costly than an alternative service or sequence of services at least as likely to produce equivalent therapeutic or diagnostic results as to the diagnosis or treatment of that patient’s illness, injury or disease.