Immunotherapy and
cell therapy as a practical approach in cancer therapy
Sogand Vahidi 1, Seyedeh Elham Norollahi 2, Ali Akbar Samadani 3,4 *
1 Medical Biology Research Center, Kermanshah University of Medical
Sciences, Kermanshah, Iran
2 Cancer Research Center and
Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
3 Department of Basic Medical
Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
4 Guilan Road Trauma
Research Center, Guilan University of Medical Sciences, Rasht, Iran
*Corresponding
Author: Ali Akbar Samadani
* Email: a_a_hormoz@yahoo.com
Abstract
Cancer treatment is one of the most important challenges in medical
science. Most methods such as surgery, chemotherapy, and radiation therapy are
not completely effective in treating cancer. In this way, immunotherapy and cell therapy have
revolutionized cancer treatment. Immunotherapy and cell therapy, like
chemotherapy, are given systemically and are effective in preventing the spread
of malignancies, but in contrast, they only attack malignant cells and have
little effect on their cells, and are more specific. Slowly They have
completely different efficiencies depending on the different types of
immunotherapy, which include selective cell transfer (ACT) and
immunosuppressive inhibitors (ICIs). The study of the mechanisms underlying the
escape of cancer cells from the immune system is also very important in
identifying new cancer treatments. This review discusses the types of
immunotherapy and cell therapy in cancer, the history of development, and
recent findings on the penetration of immune cells into the tumor and its
relationship to cancer immunotherapy, focusing on new studies and its potential
clinical applications.
Keywords: Cancer immunotherapy, Immune system, Tumor, Therapy
Introduction
Cancer,
the most dangerous disease, kills millions of patients. Comprehensive knowledge
of cancer biology allows researchers to design more appropriate treatment
systems. The type of treatment depends on the type of cancer and the progress
and purpose of the treatment. Surgery is the first option for the direct
removal of tumors in an area. Radiotherapy destroys tumors by damaging the DNA
of cancer cells. Chemotherapy helps reduce or stop the growth of tumors by
using highly toxic drugs. Immunotherapy involves the use of monoclonal
antibodies, cancer vaccines, and acceptor cell transfusions. It has now become
an important treatment for cancer with acceptable clinical results (1, 2). In addition, stem cell therapy has
provided a promising option in the fight against cancer (3). Due to the increased targeting of
tumors, the therapeutic effect of other treatments has improved. Many stem
cell-based strategies have already been clinically tested and have great
achievements and challenges in the field of cancer. Therefore, further evaluation
is needed to prepare these methods for clinical trials (4, 5). In addition, in the event of
persistent or recurrent disease, a small number of treatment strategies can
eliminate the remaining malignant cells and treatments that are more effective.
There is much evidence to support the vital role of the immune system,
especially lymphocytes, in controlling and eradicating cancer. Inhibiting the
immune system to achieve clinical efficacy has been the focus of many
treatments (6, 7). More than two decades have passed
since Gross and colleagues demonstrated the principle of genetic alteration of
cytotoxic T lymphocytes into tumor cells, and have done their main work by
stating that chimeric T cell receptors with anti-tumor properties can test this
method in the fight against tumors. Provides humanity, passes (8). On the other hand, in the last
decade, the role of the immune system in controlling tumorigenesis and tumor
progression has been well established. Although the role of adaptive immune
responses (e.g., mediated by lymphocytes) has been widely expressed, the
function of innate immune responses is less well known. The collected evidence
shows a correlation between tumor permeable lymphocytes (TIL) in cancerous
tissue and favorable prognosis in malignant types (9, 10).
A
key factor in the limited response seen in various experiments is the
complexity of immune-host tumor interactions and the existence of several
redundant tumor-mediated immune suppression mechanisms. It is also essential to
have a thorough understanding of the principles of tumor antigen
production/maintenance, antigenic evolution, and tumor immunity heterogeneity.
Further efforts in basic research should also clarify the structure and
function of most regulatory immune pathways and their specific role in various
human malignancies (11).
Understanding
the dynamic interactions between tumor cells and the immune system allows us to
specialize in immune therapies and design optimal combination approaches to
improve outcomes in patients with advanced malignancy. In a study by Samadani
et al. 2020, the main programs optimized for the treatment of CAR-T cancer
cells were classified and reviewed. The results showed that immunotherapy of
cancer by CAR-T cells (chimeric antigen receptor (CAR) T-cell therapy) has
significant clinical potential in malignancies. In other words, it is a good
cancer treatment. CAR is a specific recombinant protein compound that targets
the structure of antibodies to activate T cells. CAR-T cells can kill B cell
malignancies (12). Therefore, research on the risks
as well as strategies to neutralize the possible consequences of the tumor is
of great importance, as successful protocols and strategies in the treatment of
CAR-T cells can improve the efficiency and safety of cancers (Table 1).
Table1. CAR T-cell therapies
accepted by FDA.
Targeted antigen |
Name |
Target disease |
Patients |
BCMA |
Ciltacabtagene
autoleucel |
Multiple
myeloma |
Adults with
multiple myeloma |
Idecabtagene vicleucel |
Multiple myeloma |
Adults with multiple myeloma |
|
CD19 |
Tisagenlecleucel |
B cell
non-Hodgkin's lymphoma (NHL) / B cell acute lymphoblastic leukemia |
Children and
young adults with B cell acute lymphoblastic leukemia |
Brexucabtagene autoleucel |
B cell non-Hodgkin's lymphoma
(NHL) |
Adults with B cell non-Hodgkin's
lymphoma (NHL) |
|
Lisocabtagene
maraleucel |
B cell
non-Hodgkin's lymphoma (NHL) |
Adults with B
cell non-Hodgkin's lymphoma (NHL) |
|
Axicabtagene ciloleucel |
Follicular lymphoma /
Non-Hodgkin's lymphoma (NHL) |
Adults with follicular
lymphoma and NHL |
A
study by Chu in 2020 examined the mechanisms for using different types of stem
cells in the treatment of cancer. Recent advances in the clinical applications
of stem cells as well as the common risks of this treatment have been
summarized. Depending on their inherent capacities, different types of stem
cells have been used for anti-cancer treatment. HSC transplantation has
provided an effective treatment for cancers such as leukemia, multiple myeloma
and lymphoma. Simultaneous injection of mesenchymal stem cells leads to
modulation of the immune system, high effects in reducing cases of
graft-versus-host disease (GVHD) and also repair of damaged tissues after heavy
chemotherapy or radiotherapy. The defined induction protocol enables the
production of a large number of clinically world-class immune cells for further
evaluation in humans. Finally, targeting CSCs with the delivery of CSC and PSC
antigen-derived anticancer vaccines may provide promising strategies to prevent
tumor growth, metastasis, and recurrence. As a result of this research, to
improve the overall results in the fight against cancer, guidelines for the
future are presented (13).
In a
study by Miliotou et al. 2018, concerning several conventional cytotoxic
methods for neoplastic diseases and their limited effectiveness according to
the heterogeneity of cancer cells, a study was conducted for better treatment
approaches such as immunotherapy. Increases the patient's immunity. CAR-T cell
therapy involves the genetic modification of a patient's autologous T cells to
express a specific CAR for a tumor antigen (Figure 1). Clinical trials have
shown very promising results in patients in the final stage with a complete
recovery of up to 92% in acute lymphocytic leukemia (14).
In a
study by Saadatpour et al. in 2017, they examined cell therapy and gene therapy
and imaging techniques. Studies have shown that cancer is one of the most
important health problems in the world and has created many challenges; the
current understanding of this disease is very useful in the emergence of a
number of new treatments. Among these, cell therapy and gene therapy are known
as new and effective therapies. One of the major challenges of cell therapy and
gene therapy in cancer is the proper monitoring of modified cells and genes.
Visual tracking of cell therapy, immune cells, stem cells and genetic vectors
containing therapeutic genes and various drugs is important in the treatment of
cancer. Similarly, molecular imaging such as nanosystems, fluorescence,
bioluminescence, positron emission tomography, and photon emission computed
tomography has also been identified as powerful tools in monitoring cancer
patients receiving cell therapy and gene therapy or drugs (15).
A
2016 study by Velcheti et al. Focused on the basic principles of immunotherapy,
new pathways, and hybrid immunotherapy. This study showed that the recent
success of immunotherapy strategies such as blockade of immunosuppression in
several malignancies has highlighted the role of immunotherapy in the treatment
of cancer. Cancers use several mechanisms to select host-tumor immune
interactions that lead to escape from the immune system. Our understanding of
host tumor interactions has evolved over the past few years and has led to promising
new treatment strategies (16).
Figure 1. General mechanism of CAR T cells
therapy.
A
2016 study by Velcheti et al. Focused on the basic principles of immunotherapy,
new pathways, and hybrid immunotherapy. This study showed that the recent
success of immunotherapy strategies such as blockade of immunosuppression in
several malignancies has highlighted the role of immunotherapy in the treatment
of cancer. Cancers use several mechanisms to select host-tumor immune
interactions that lead to escape from the immune system. Our understanding of
host tumor interactions has evolved over the past few years and has led to promising
new treatment strategies (16).
The
occurrence of molecular remission after donor lymphocyte infusion (DLI) in
myeloid tumours relapsing after bone marrow transplantation was the first
demonstration of the usefulness of adoptive T cell therapy (ATC) in human
cancers (17). Increasingly broad tumor infiltrating
lymphocytes (TIL) have also been shown to implementation details and long-term
transformation of large vascularized metastatic melanomas (18). In numerous EBV-associated cancers, such
as Hodgkin's disease, Burkitt's lymphoma, and nasopharyngeal carcinoma, ATC
utilizing Epstein-Barr virus-specific T cells demonstrated clinical benefit (19). Furthermore, once circulating
tumor-reactive T cells from a patient's peripheral blood were ex vivo increased
in acceptable numbers and administered to the patients, therapeutic efficacy
was observed (20). Whereas these treatments depend on
endogenous T cell repertoires, technical developments in T cell engineering
with retroviral and plasmid vectors have enabled the genetic introduction of
tumor specific T cell receptors (TCRs) or CARs to generate large numbers of
tumor targeting T cells. Unlike TCRs, that either acknowledge peptides obtained
from cellular proteins introduced in the context of the major
histocompatibility complex (MHC), CARs identify any surface antigen, such as
carbohydrates and phospholipids, with elevated MHC independent identification.
Hematological malignant tumors are the focus of about 65 percent of the
research (21). Whereas CD19 is the most commonly
targeted antigen in hematological B-cell tumours (>80%), research is ongoing
to look into other antigens also including ROR1, B-cell maturation antigen,
CD20, CD22, CD30, CD33, CD123, CD133 and CD138 (22). Patients with various B-cell cancers
have been considered with CD19 CAR T cells in research trials (Figure 2), and
while solid tumors were the first targets of CAR T cell treatments (23, 24){Maus, 2014
#45}, realistic
clinical reactions have been seen in clinical studies where patients with
various B-cell carcinomas have been managed with CD19 CAR T cells.
Figure 2.
Components elaborate in TCR and CAR identification and activation. TCR consists
of an α and a β chain, which is expressed in complex with CD3 chains. TCR
detects proteins presented as peptides by MHC molecules. Stimulation of CD28 is
essential for the activation and generation of interleukin-2 and other
cytokines.Ttumor cells as well as antigen-presenting cells in the tumor
microenvironment do not express excitatory molecules. While first-generation
CARs signal only through the CD3ζ chain, second-generation CARs contain a
signaling amplitude of an excitatory molecule, e.g., CD28.
Among the first hopeful findings in the context from
National Cancer Institute (NCI) researchers, which released a case analysis in
2010 in which a heavily pretreated patient with follicular lymphoma experienced
a dramatic partial remission (PR) after obtaining preconditioning chemotherapy
obeyed by intake of T cells retrovirally transduced to describe a
second-generation CD19 CAR with CD28 costimulation parts. T cells induced with
a lentiviral vector transporting a CD19 CAR with a 4-1BB costimulation parts
displayed remarkable antileukemia usefulness (25, 26). Two of the three treated groups with end-stage
developed chronic lymphocytic leukemia (CLL) achieved complete remissions (CR),
with the third achieving a limited response. The outcomes of the experiment
after full enrollment were recently released, and they showed an increased
frequency of 57 percent, with 4 of 14 treated patients in CR and 4 PRs (27). Interestingly, the research found that
the knowledge of CAR T cells to develop in vivo over time is associated with
clinical outcomes. Moreover, CAR T cells survived and functioned for more than
four years in the first two patients who achieved CR with no recurrence. Even
before merged with ibrutinib, a small-molecular inhibitor of the enzyme
Bruton's tyrosine kinase (BTK) correlated with greater B-cell activation and
expansion, CD19 CAR T cell role and engraftment may be enhanced additional (28).
One of the most impressive reactions with CD19
redirected T cells have already been noted by organizations at UPENN, Memorial
Sloan Kettering Cancer Center (MSKCC), and the National Cancer Institute (NCI)
in patients with protective or recurred acute lymphoblastic leukemia (ALL),
with CR rates from 70 to 90 cases in around 65 cases across the three tests (29). Whereas the therapy affected the
cytogenetic and molecular treatment of her leukemia, a second BMT administered
three months after the T cell injection precludes an interpretation of the
UCART19 therapy's long-term effectiveness. To prevent alloreactivity, the
infused allogeneic CAR-expressing T cells were gene transcribed with nucleases
to interrupt the expression of the endogenous TCRs (30).
Allogeneic CAR T cells have been shown in research to
not only stimulate tumorigenesis but rather to drive GVHD, and as such the
UCART19 method is analytically essential for high TCR silencing effectiveness
or efficient depletion of TCR expressing T cells prior to infusion. A clinical
study of a multiple myeloma patient in remission after CD19 CAR T cell
treatment is also noteworthy, despite the lack of noticable CD19 expression in
99.95 percent of the patient's neoplastic plasma cells (31, 32). It is theorised that the reaction is
induced by that of the abolition of a small population of CD19 expressing
myeloma stem cells or the elimination of CD19 expressing cells that play a
major role in the maintenance of myeloma biogenesis. In patients with different
chemotherapy-refractory B-cell lymphomas, inspire and motivate clinical
outcomes have been acquired, such as CR in four out of seven clinically
important patients with diffuse large B-cell lymphoma (DLBCL) after infusion of
CD19 CAR T cells (33). In comparison, the clinical utility of
CAR T cells in nonhematological, tumor cells has been hard to demonstrate. CARs
targeting mesothelin, which would be upregulated in human epidermal growth
factor receptor family members, a variety of solid tumors, upregulated in pancreatic, breast,
non-small-cell lung cancer, bladder, neuroblastoma associated GD2, salivary
gland, endometrial, and ovarian predominate among open clinical
guidelines. Nevertheless, a number of
targets are being studied in clinical studies, including MUC1 and
carcinoembryonic antigen (CEA), which are upregulated in numerous malignancies,
vascular endothelial growth factor receptor 2 (VEGFR2), which is upregulated in
tumor vasculature and fibroblast activation protein (FAP), which targets
cancer-associated fibroblasts in the tumor stroma (34-36).
Conclusion
Adoptive transfer of gene-modified T cells is a novel
treatment method that is still in its early stages. CAR-redirected T cells are
renewable medicine that can proliferate in the patient after infusion and then
persist and provide develop a system immunity. Clinical efficacy with CD19 CAR
T cells in leukemia and lymphoma has enhanced the field and resulted in
substantial pharmaceutical and venture investment capital in the biotech
segment, and also developing successful academic-industrial partnerships to
investigate new findings in basic research that may translate into clinical and
commercial effect.
This quickly evolving area difficulties
that must be acknowledged in order to realize the promise of CAR T cell therapy
for a broader use. Whereas CAR T cell treatments have shown promising
preliminary efficacy in solid tumors, clinical data to date has fallen far
short of expectations for game-changing cell therapy. Translational study is
highly concerned with improving the specificity, usefulness, and safety of CAR
T cells for use in cancers other than leukemia. Truly tumor-specific surface
antigens are rare, and effective processes to mitigate life-threatening and
unexpected off-target toxicities are critical. T cell therapies together with
immunomodulatory agents such as checkpoint inhibitors and cytokines, as well as
small-molecular antagonists which prevent biological reactions important for
tumorigenesis, represent an exciting opportunity that may have a synergic
effect in augmenting antitumor reactions.Advancements in genetic manipulation ,
T cell choice and expansion strategies, and the development of safer and more
effective viral and nonviral vectors will all help to improve T cell gene
therapy integration. Finally, significant efforts are being made to develop
universal and off-the-shelf, allogeneic T cell drugs in order to overcome the
difficulties in dealing with complicated logistics and production of
individualized T cell treatment in the autologous configuration.
Author contributions
SV and SEN wrote and completed the manuscript. AAS
designed, wrote and edited the manuscript comprehensively. All authors
confirmed the final version of the paper.
Conflict of interest
The authors declare that they have no conflicts of interest.
References
1. Vasievich
EA, Huang L. The suppressive tumor microenvironment: a challenge in cancer
immunotherapy. Mol Pharm. 2011;8(3):635-41.
2. Vanneman M, Dranoff G.
Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev
Cancer. 2012;12(4):237-51.
3. Young A, Quandt Z, Bluestone
JA. The balancing act between cancer immunity and autoimmunity in response to
immunotherapy. Cancer Immunol Res. 2018;6(12):1445-52.
4. Gomes JP, Assoni AF, Pelatti
M, Coatti G, Okamoto OK, Zatz M. Deepening a simple question: Can MSCs be used
to treat cancer? Anticancer Res. 2017;37(9):4747-58.
5. Samadani AA, Keymoradzdeh A,
Shams S, Soleymanpour A, Norollahi SE, Vahidi S, et al. Mechanisms of cancer
stem cell therapy. Clin Chim Acta. 2020;510:581-92.
6. Almåsbak H, Aarvak T, Vemuri
MC. CAR T cell therapy: a game changer in cancer treatment. J Immunol Res.
2016;2016.
7. Norollahi SE, Mansour-Ghanaei
F, Joukar F, Ghadarjani S, Mojtahedi K, Nejad KG, et al. Therapeutic approach
of Cancer stem cells (CSCs) in gastric adenocarcinoma; DNA methyltransferases
enzymes in cancer targeted therapy. Biomed Pharmacother. 2019;115:108958.
8. Gross G, Waks T, Eshhar Z.
Expression of immunoglobulin-T-cell receptor chimeric molecules as functional
receptors with antibody-type specificity. Proc Natl Acad Sci.
1989;86(24):10024-8.
9. Schalper KA, Brown J,
Carvajal-Hausdorf D, McLaughlin J, Velcheti V, Syrigos KN, et al. Objective
measurement and clinical significance of TILs in non–small cell lung cancer. J
Natl Cancer Inst. 2015;107(3):dju435.
10. Velcheti V, Schalper KA,
Carvajal DE, Anagnostou VK, Syrigos KN, Sznol M, et al. Programmed death
ligand-1 expression in non-small cell lung cancer. Lab Invest.
2014;94(1):107-16.
11. Thaxton JE, Li Z. To affinity
and beyond: harnessing the T cell receptor for cancer immunotherapy. Hum
Vaccines Immunother. 2014;10(11):3313-21.
12. Samadani AA, Keymoradzdeh A,
Shams S, Soleymanpour A, Rashidy-Pour A, Hashemian H, et al. CAR T-cells
profiling in carcinogenesis and tumorigenesis: an overview of CAR T-cells
cancer therapy. Int Immunopharmacol. 2021;90:107201.
13. Chu D-T, Nguyen TT, Tien NLB,
Tran D-K, Jeong J-H, Anh PG, et al. Recent progress of stem cell therapy in
cancer treatment: molecular mechanisms and potential applications. Cell.
2020;9(3):563.
14. Miliotou AN, Papadopoulou LC.
CAR T-cell therapy: a new era in cancer immunotherapy. Curr Pharm Biotechnol.
2018;19(1):5-18.
15. Saadatpour Z, Rezaei A,
Ebrahimnejad H, Baghaei B, Bjorklund G, Chartrand M, et al. Imaging techniques:
new avenues in cancer gene and cell therapy. Cancer Gene Ther. 2017;24(1):1-5.
16. Velcheti V, Schalper K. Basic
overview of current immunotherapy approaches in cancer. Am Soc Clin Oncol Educ
Book. 2016;36:298-308.
17. Deol A, Lum LG. Role of donor
lymphocyte infusions in relapsed hematological malignancies after stem cell transplantation
revisited. Cancer Treat Rev. 2010;36(7):528-38.
18. Hinrichs CS, Rosenberg SA.
Exploiting the curative potential of adoptive T‐cell therapy for cancer.
Immunol Rev. 2014;257(1):56-71.
19. Heslop HE, Slobod KS, Pule MA,
Hale GA, Rousseau A, Smith CA, et al. Long-term outcome of EBV-specific T-cell
infusions to prevent or treat EBV-related lymphoproliferative disease in
transplant recipients. Am J Hematol. 2010;115(5):925-35.
20. Yee C, Lizee G, Schueneman AJ.
Endogenous T-cell therapy: clinical experience. Cancer J. 2015;21(6):492-500.
21. Gill S, June CH. Going viral:
chimeric antigen receptor T‐cell therapy for hematological malignancies.
Immunol Rev. 2015;263(1):68-89.
22. Berger C, Sommermeyer D,
Hudecek M, Berger M, Balakrishnan A, Paszkiewicz PJ, et al. Safety of targeting
ROR1 in primates with chimeric antigen receptor–modified T cells. Cancer
Immunol Res. 2015;3(2):206-16.
23. Carpenter RO, Evbuomwan MO,
Pittaluga S, Rose JJ, Raffeld M, Yang S, et al. B-cell maturation antigen is a
promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer
Res. 2013;19(8):2048-60.
24. Maus MV, Grupp SA, Porter DL,
June CH. Antibody-modified T cells: CARs take the front seat for hematologic
malignancies. Am Soc Hematol 2014;123(17):2625-35.
25. Kalos M, Levine BL, Porter DL,
Katz S, Grupp SA, Bagg A, et al. T cells with chimeric antigen receptors have
potent antitumor effects and can establish memory in patients with advanced
leukemia. Sci Transl Med. 2011;3(95):95ra73-95ra73.
26. Porter DL, Levine BL, Kalos M,
Bagg A, June CH. Chimeric antigen receptor–modified T cells in chronic lymphoid
leukemia. N engl j Med. 2011;365:725-33.
27. Porter DL, Hwang W-T, Frey NV,
Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen receptor T cells persist
and induce sustained remissions in relapsed refractory chronic lymphocytic
leukemia. Sci Transl Med. 2015;7(303):303ra139-303ra139.
28. Fraietta JA, Beckwith KA, Patel
PR, Ruella M, Zheng Z, Barrett DM, et al. Ibrutinib enhances chimeric antigen
receptor T-cell engraftment and efficacy in leukemia. Blood
2016;127(9):1117-27.
29. Lee DW, Kochenderfer JN,
Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing
CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children
and young adults: a phase 1 dose-escalation trial. Lancet.
2015;385(9967):517-28.
30. Poirot L, Philip B,
Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, et al. Multiplex
genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive T-cell
immunotherapies. Cancer research. 2015;75(18):3853-64.
31. Jacoby E, Yang Y, Qin H, Chien
CD, Kochenderfer JN, Fry TJ. Murine allogeneic CD19 CAR T cells harbor potent
antileukemic activity but have the potential to mediate lethal GVHD. Blood.
2016;127(10):1361-70.
32. Garfall AL, Maus MV, Hwang W-T,
Lacey SF, Mahnke YD, Melenhorst JJ, et al. Chimeric antigen receptor T cells
against CD19 for multiple myeloma. N Engl J Med. 2015;373(11):1040-7.
33. Kochenderfer JN, Dudley ME,
Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M, et al.
Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell
malignancies can be effectively treated with autologous T cells expressing an
anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015;33(6):540.
34. Whilding LM, Maher J.
ErbB-targeted CAR T-cell immunotherapy of cancer. Immunotherapy.
2015;7(3):229-41.
35. Feng K, Guo Y, Dai H, Wang Y,
Li X, Jia H, et al. Chimeric antigen receptor-modified T cells for the
immunotherapy of patients with EGFR-expressing advanced relapsed/refractory
non-small cell lung cancer. Sci China Life Sci. 2016;59(5):468-79.
36. You F, Jiang L, Zhang B, Lu Q,
Zhou Q, Liao X, et al. Phase 1 clinical trial demonstrated that MUC1 positive
metastatic seminal vesicle cancer can be effectively eradicated by modified
Anti-MUC1 chimeric antigen receptor transduced T cells. Sci China Life Sci.
2016;59(4):386-97.