Adoptive Cell Therapy of Gastric Cancer


Engineered T cells

Target antigen

Cancer

Number of patients treated in trial

Results

Reference

Year reported

TCR-T

gp100

Melanoma

Sixteen

One CR and two PR

[29]

2009

MART-1/Melan-A

Melanoma

Thirty-one

Four OR

[28, 62]

2006, 2009

Twenty

Six PR

[29]

2009

p53

Melanoma

Fourteen

One PR

[63]

2010

NY-ESO-1

Melanoma and synovial sarcoma

Seventeen

Two CR and seven PR

[30]

2011

Synovial cell sarcomas

Eighteen

Eleven RR

[31]

2015

Melanoma

Twenty

Eleven RR

[31]

2015

Multiple myeloma

Twenty

Sixteen RR

[32]

2015

CEA

Colorectal

Three

One PR

[64]

2011

MAGE-A3

Melanoma, esophageal and synovial sarcoma

Nine

One CR and four PR

[65]

2013

Melanoma and MM

Two

Lethal cardiac toxicity

[35]

2013

CAR-T

CD19

CLL

Three

Two CR and one PR

[44, 66]

2011

Lymphoma and CLL

Seven

One CR, five PR, and one SD

[42, 67]

2012, 2010

ALL

Sixteen

Fourteen CR

[45]

2014

Pediatric and adult ALLs

Thirty

Twenty-seven CR

[39]

2014

NHL

Six

Two SD to 10 months

[68]

2011

CD20

NHL and mantle cell lymphoma

Seven

Two CR, one PR, four SD

[69]

2008

NHL

Three

One PR, two NED maintained

[70]

2012

CD171

Neuroblastoma

Six

One PR

[71]

2007

GD2

Neuroblastoma

Nineteen

Three CR

[50]

2011

ERBB2

HNSCC

Proposed

[72]

2013

Colorectal cancer

One

Died of respiratory distress

[48]

2010

Sarcoma

Seventeen

Four SD

[59]

2015

CEA

Colorectal and breast cancer

Seven

Two minor response

[73]

2002
 
Gastrointestinal cancer

Nine

One SD

[74]

2015

Lewis Y

AML

Four

One cytogenetic remission

[75]

2013
 
CAIX

Renal cell carcinoma

Twelve

No clinical response

[49]

2013


Abbreviations: MART-1 melanoma antigen recognized by T cells 1, Melan-A melanocyte antigen, MAGE-A3 melanoma-associated antigen 3, CEA carcinoembryonic antigen, NY-ESO-1 New York esophageal squamous cell carcinoma-1, ALL acute lymphoblastic leukemia, CLL chronic lymphocytic leukemia, MM multiple myeloma, ERBB2 erythroblastosis oncogene B2, GD2 ganglioside, CAIX carbonic anhydrase-IX, NHL non-Hodgkin’s lymphoma, HNSCC head and neck squamous cell carcinoma, CR complete response, PR partial response, OR objective response, SD stable disease, RR response rate, NED no evidence of disease






11.6 Conclusions and Future Perspectives


According the remarkable clinical trial results described here, we strongly believe that ACT, including TIL, CTL, TCR-T, and CAR-T transfer therapy, is the most promising “living” treatment for targeting human tumors, in which T cells are the terminator. However, there remains a need to improve the killing ability of transfer cells, while reducing the side effects of ACT, such as on-target/off-tumor toxicities, cytokine release syndrome, and, at the same time, develop an ability to harness the immune microenvironment, in order to make ACT an even more successful treatment option.

In order to prevent the on-target/off-tumor toxicities, target antigens that are only expressed on tumor cells, and not on normal tissue cells, must be selected. The tumor-specific “nonself” immunogenic neoantigens encoded by either viral genes or through somatic mutations possess the potential to induce specific anticancer immunity, including cellular and humoral immune responses. Today, numerous clinical trials demonstrate that although these “nonself” antigens initiate the antigen-specific immunoglobulin G antibodies and CD4+/CD8+ T cells response, not all of them show a clinical benefit in the response rate, progression-free survival, or overall survival [7678].

Personalized cell therapy is the key to cure human malignant diseases. There are five steps required to reach this goal. First, cancer mutations must be identified through exome sequencing. Second, tandem minigenes or synthetic peptides of all identified mutations must be created and introduced to autologous APC. Third, mature autologous APCs coculture with T cells form peripheral blood or tumor. Fourth, tumor-reactive T cells must accumulate together through 4-1BB or OX40 positive selection. Fifth, rapid expansion of such cells in vitro must be carried out and used to treat the tumor in vivo, or a PCR of the TCR of such cells must be carried out for TCR-T treatment (Figs. 11.1 and 11.2) [26].

A372008_1_En_11_Fig1_HTML.jpg


Fig. 11.1
A “blueprint” for T cells treatment targeting tumor-specific mutations (Adapted from Adoptive cell transfer as personalized immunotherapy for human cancer (2015), Rosenberg SA, Restifo NP, [26])


A372008_1_En_11_Fig2_HTML.gif


Fig. 11.2
Gene-engineered peripheral blood lymphocytes (Adapted from Adoptive cell transfer as personalized immunotherapy for human cancer (2015), Rosenberg SA, Restifo NP, [26])

Moreover, there exists accumulating correlative data which suggests that directly sorting PD-1+ lymphocytes in peripheral blood could function as an alternative noninvasive strategy to develop neoantigen-reactive lymphocytes or TCRs to treat melanomas (Fig. 11.3) [79].

A372008_1_En_11_Fig3_HTML.gif


Fig. 11.3
A new strategy for generating autologous TCR gene treatments targeting neoantigens of advanced epithelial cancers (Adapted from Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients (2016), Gros A et al. [79])

Gastric cancer tumors have been shown to exhibit a high prevalence of mutations, with many occurring with EB virus infection. Both mutation antigens and viral antigens are ideal antigen targets for gastric cancer immunotherapy. Thus, we can use the abovementioned strategies to prepare T cells in clinical ACT for gastric cancer.


References



1.

Schmidt-Wolf IG, Negrin RS, Kiem HP, Blume KG, Weissman IL. Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med. 1991;174(1):139–49.CrossRefPubMed


2.

Shi L, Zhou Q, Wu J, Ji M, Li G, Jiang 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. doi:10.​1007/​s00262-012-1289-2.CrossRefPubMedPubMedCentral


3.

Liu K, Song G, Hu X, Zhou Y, Li Y, Chen Q, et al. A positive role of cytokine-induced killer cell therapy on gastric cancer therapy in a chinese population: a systematic meta-analysis. Med Sci Monit. 2015;21:3363–70.CrossRefPubMedPubMedCentral


4.

Li X, Dai D, Song X, Liu J, Zhu L, Xu W. A meta-analysis of cytokine-induced killer cells therapy in combination with minimally invasive treatment for hepatocellular carcinoma. Clin Res Hepatol Gastroenterol. 2014;38(5):583–91. doi:10.​1016/​j.​clinre.​2014.​04.​010.CrossRefPubMed


5.

Wang ZX, Cao JX, Liu ZP, Cui YX, Li CY, Li D, et al. Combination of chemotherapy and immunotherapy for colon cancer in China: a meta-analysis. World J Gastroenterol. 2014;20(4):1095–106. doi:10.​3748/​wjg.​v20.​i4.​1095.CrossRefPubMedPubMedCentral


6.

Han RX, Liu X, Pan P, Jia YJ, Yu JC. Effectiveness and safety of chemotherapy combined with dendritic cells co-cultured with cytokine-induced killer cells in the treatment of advanced non-small-cell lung cancer: a systematic review and meta-analysis. PLoS One. 2014;9(9):e108958. doi:10.​1371/​journal.​pone.​0108958.CrossRefPubMedPubMedCentral


7.

Jiang J, Xu N, Wu C, Deng H, Lu M, Li M, et al. Treatment of advanced gastric cancer by chemotherapy combined with autologous cytokine-induced killer cells. Anticancer Res. 2006;26(3B):2237–42.PubMed


8.

Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 1986;233(4770):1318–21.CrossRefPubMed


9.

Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988;319(25):1676–80. doi:10.​1056/​NEJM198812223192​527.CrossRefPubMed


10.

Feldman SA, Assadipour Y, Kriley I, Goff SL, Rosenberg SA. Adoptive cell therapy—tumor-infiltrating lymphocytes, T-cell receptors, and chimeric antigen receptors. Semin Oncol. 2015;42(4):626–39. doi:10.​1053/​j.​seminoncol.​2015.​05.​005.CrossRefPubMed


11.

Andersen R, Donia M, Westergaard MC, Pedersen M, Hansen M, Svane IM. Tumor infiltrating lymphocyte therapy for ovarian cancer and renal cell carcinoma. Hum Vaccin Immunother. 2015;11(12):2790–5. doi:10.​1080/​21645515.​2015.​1075106.CrossRefPubMedPubMedCentral


12.

Turcotte S, Gros A, Hogan K, Tran E, Hinrichs CS, Wunderlich JR, et al. Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy. J Immunol. 2013;191(5):2217–25. doi:10.​4049/​jimmunol.​1300538.CrossRefPubMedPubMedCentral

Jun 26, 2017 | Posted by in ONCOLOGY | Comments Off on Adoptive Cell Therapy of Gastric Cancer

Full access? Get Clinical Tree

Get Clinical Tree app for offline access