SlideShare une entreprise Scribd logo
1  sur  8
Télécharger pour lire hors ligne
Luke Brennan
4/21/2015
Review of Adoptive T-Cell Immunotherapy
INTRODUCTION
One of the ‘hallmarks’ of cancer is its ability to evade detection and destruction by the body’s
immune system by essentially hiding in plain sight1
. Cancer is also inherently derived from the body’s
own tissue, and harbors many of the signals that identify it as ‘self,’ which contributes to its
camoflauge1
. These two factors have made cancer so difficult to treat, because the immune system is
insufficient and man-made chemicals and radiation therapy used to attack cancer inevitably targets
unintended healthy tissue.
These issues and more prompted the development of immunotherapy for cancer treatment.
Much like how a vaccination conditions the adaptive immune system to respond to a certain pathogen,
immunotherapy would allow the immune system to sense and destroy cancer, though it uses different
means. This is often referred to as the cancer immunosurveillance hypothesis, and it gained significant
support after the verification of human tumor-associated antigens (TAAs) which are identifiers unique to
cancer, and can be exploited to discern cancer from healthy tissue2
.
Although immunotherapy is used to treat cancer in many ways, the scope of this paper will be
limited to Adoptive T-Cell Therapy (ACT). T-cells are lymphocytes, or white blood cells, which are present
throughout the body and attack pathogens and cells marked for apoptosis. T cells usually destroy cells
that would become cancerous before they even become a problem, when these mutated cells aren’t
destroyed and proliferate in an uncontrolled way they become cancers. ACT is the process by which T-
cells (originally extracted from the body, in most cases and for the purposes of this paper) with cancer-
recognizing properties are introduced into the patient to combat cancer.
Harnessing the power of these cancer sensitive T cells for use against cancer has great potential
for many reasons:
“1) T cell responses are specific, and can thus potentially distinguish between healthy
and cancerous tissue;
2) T cells responses are robust, undergoing up to 1,000-fold clonal expansion after activation;
3) T cell response can traffic to the site of antigen, suggesting a mechanism for eradication of
distant metastases;
4) T cell responses have memory, maintaining therapeutic effect for many years after initial
treatment.”2
As elegant a solution as immunotherapy may seem for the above reasons, many challenges
must be overcome before this becomes a viable treatment for mainstream cancer patients, as discussed
later in this review. Regardless, the following sections describe two main types of ACT based on the anti-
tumor activity of the lymphocytes collected from the patient.
ACT USING TUMOR-INFILTRATING LYMPHOCYTES
This type of ACT is dependent on the collection of lymphocytes with anti-tumor cytotoxic
activity, called tumor-infiltrating lymphocytes (TILs), against the cancer in question2
. Fortunately, such T-
cells have been identified in the tumor samples of up to 80% of melanoma patients, with admittedly
lower frequency in other forms of cancer5
. In fact, though not universal, lymphocyte infiltration of tumor
tissue has in fact become a hallmark of cancer1
. In the cases where such cells are detected and can be
extracted, these TILs can be cultured and grown ex vivo.
This process begins with the TILs from a “micro culture derived from a single tumor fragment or
106
viable cells derived from a single-cell enzymatic digestion [of a resected tumor specimen]”29
. These
cells are grown and cultures split as needed for two weeks with high dose interleukin 2, after which
point each culture is kept at 0.8-1.6x106
mL-1
in flasks and generates roughly 5x107
cells (the required
minimum being 3x107
cells) from each culture after 3-5 weeks29
. The cultures are then tested for activity
and specificity by an immunosorbent assay after stimulation with tumor cells, and cultures deemed
active undergo a rapid expansion phase for 2 weeks with donor feeder cells, anti-CD3 OKT-3 monoclonal
antibody, and high dose interleukin 2 to promote expansion 29
. In total, the entire manufacture process
takes roughly 6-8 weeks31
. The expanded colonies of TILs are then reintroduced to the body to attack
the cancer with greater numbers than before. This approach capitalizes on the ‘strength in numbers’
strategy, but with the added benefit of more specific TILs from the testing and selection processes
performed ex-vivo29
. This type of ACT would be considered the most feasible, but it sacrifices efficacy.
ACTs using TILs are limited in a few ways. First, this treatment is simply not an option for any
patient in whom TILs cannot be identified or extracted. Even for those patients from whom TILs can be
extracted, there is growing concern that these TILs may have little to no effect on tumor progression
despite their presence in areas of tumor inhibition2
. Further, to avoid unintended autoimmune attack,
these TILs generally have low affinity to the self-antigens and germline antigens that are usually
overexpressed by cancerous tissues (but are shared by healthy body tissues)6
. While this is still the most
effective method of ACT to date, these limitations have encouraged development of the second type of
ACT.
ACT USING ENGINEERED T CELLS
This method of ACT makes use of the T-cells’ time ex vivo (as described in the methods section)
to enhance T-cell function by changing receptors to give the cells improved affinity to self-antigens or
novel specificity, and/or by improving their proliferative capacity through alterations in signaling
functions7
. Because the cells are being engineered, the T-cells do not necessarily need to be TILs or even
from the tumor region, and can be collected from peripheral blood samples which are much easier to
collect, and are present in every patient8
. This higher affinity and novel specificity is largely achieved
through expression of heterodimeric T-cell receptors (TCRs) in T-cells which can allow tighter binding to
the self-antigens overexpressed on cancer, or neo-antigens, which are epitopes that are only present in
cells with cancerous mutations9
. To achieve this, a gene corresponding to the engineered TCR is
introduced into the T cells by a retroviral vector32
. The TCR then “combines TCR-alpha and TCR-beta
genes” and undergoes MHC restriction, a process that ensures it only recognizes peptide antigens when
they are presented on the body’s MHC molecule10
. Another method to enhance affinity is to reduce N-
glycosylation on TCR chains11
. Surprisingly, self-antigens (specifically genes of the cancer testis family*for
more info look at 12
) rather than neoantigens, are more often the targets of such engineered TCRs12
.
Some of the first trials of this type have been for the NY ESO-1-specific and HLA-A2-restricted
TCR for use in melanoma, myeloma, and synovial cell sarcoma13
. These studies have been making great
progress and have not shown toxicity. Toxicity is a large concern in these trials, after all these cells are
being engineered to attack self-antigens that are present on healthy body tissue. In fact in another study
a TCR engineered for enhanced MAGE 3 affinity and HLA-A1-restriction had off target recognition of the
muscle protein Titin, resulting in lethal cardiac toxicity for both patients14,15
. This will be discussed
further along in the paper. Another problem of ACT with genetically engineered T-cells is that, due to a
multitude of factors known and unknown, it has been less successful than ACT with TILs16,17
. This being
said, ACT remains relevant because of how much it can improve with better protocols, and because it
greatly expands the range of ACT to cancers like neuroblastoma, synovial cell carcinoma, and colorectal
carcinoma (among others) where TILs cannot be extracted18,19, 20
.
The synthesis of these genetically engineered T-cells begins with T-cells from ficoll-purified
PMBCs, which are activated with OKT-3 antibodies29
. They are then transduced with a retroviral vector
that encodes the desired antigen-specific TCR, cultured for 2 weeks, and transduced and expanded
under cGMP 32,33
. These activated T-cells are then transduced again with retroviral vectors in
RetroNectin-coated cell bags, inoculated in a WAVE bioreactor for two days, and expanded with a
continuous perfusion regime29
. After this, the beads (from cGMP treatment) are removed and the cells
are formulated for infusion29
.
This total process takes about 2 weeks and is of a large enough scale to support multiple clinical
trials29
.
CAR T-Cells
One other noteworthy category of genetically engineered T-cells for use in ACT is the CAR-
modified T-cell. This modification exploits the fact that “the cytoplasmic tail of the TCR CD3zeta chain
[can] activate T-cells without the rest of the receptor complex”21
. Despite disappointing results from first
generation CARs22
, second and third generation CARs have shown antitumor effects and have remained
in patients for up to a decade (but also as little as a month)*for more information about the modifications of second and third
CARs go to 23
. What is promising is that response rates are generally encouraging, over 50% of patients
achieving some kind of remission in studies at the University of Pennsylvania, and that this response is
correlated with CAR T-cell proliferation, indicating that the remission is related to the presence or
activities of the CAR T-cells10
.
CAR modification represent a valuable alternative to TCR modification because only CAR-
modified T-cells can recognize a cancer cell that has lost MHC expression, a condition which applies to
many cancers24,25
. Also, CAR-modified T-cells are sometimes able to remain in the body and fight cancer
much longer than TCR-modified T-cells, though the longevity of each varies substantally26,27
. This being
said, TCR-modification has its place, as it is “able to sense the entire intracellular proteome that is
presented by MHC molecules” and accordingly can attack a wider range of molecules in the cancer
cells10
. It also requires about 10x less target antigen expression (sometimes less than 10 ligands) to
induce cytolosys28
.
DISCUSSION
ACT involving any type of T-cell is not yet ready for the standard treatment of cancers on a large
scale. This is because of several factors both biological and practical.
T-cell Inhibition
First, both the cancer and the immune system itself have several methods of inhibiting an
immune response to cancer tissue. As T-cells are maturing in the thymus, they are tested for recognition
to self-antigens, and those that are particularly reactive are deleted to avoid autoimmunity34
. These
especially active T-cells that the body stops from maturing are needed to recognize the over-expressed
self-antigens on the surface of cancer cells, so this limits the effectiveness of any TIL related ACT10
. T-
cells are also inhibited from attacking cancer in maturity by peripheral tolerance mechanisms, which
consists of 3 categories2
:
1. Inhibition from the T-cell itself or its nature, like low activity around inflammation*for more information
see 35
2. Inhibition from the Tumor, like the expression of checkpoint molecules*for more information go to 36, this is
an area of relatively high development after trials in melanoma (37,38).
3. Inhibition from regulatory T-cells and myeloid derived suppressor cells*for more information see 39
.
The first two sources of inhibition are difficult to handle, as they are meant to stop autoimmune
attack on one’s own body tissues2
. Simply turning these inhibitory mechanisms off could cause severe
autoimmune disease.
Toxicity
Toxicity is another huge concern, with off-target autoimmunity having lethal ramifications in
both TCR-modified T-cell ACT studies (see above) and CAR-modified T-cells which can cause B-cell
aplasia, lysis syndrome, and cytokine release syndrome22,40,41,10
. To make matters works, the toxicity
from these ACTs can target the brain or heart, and cannot always be managed with corticosteroids10
.
The problem here is that T-cells are being engineered to have greater recognition of self-antigens, and
while this allows them to detect the overexpressed self-antigens on cancer cells, it is difficult to
eliminate all off target effects since almost every cell in the body has these self-antigens. It goes without
saying that work is being done to stop this off target autoimmunity, and one noteworthy idea is to add
inducible mechanisms for T-cell apoptosis when engineering the T-cells ex-vivo so that they could be
stopped quickly and easily if toxicity were to become an issue10
.
Longevity
The final biological issue with ACT is response and longevity, specifically for engineered T-cells.
As noted above, most TCR-modified T-cells can often last for only a few months, and CAR-modified T-
cells aren’t much better. This is only counting the patients who respond well to the treatment. For ACT
to become a mainstream treatment, researchers need to find ways to allow the engineered cells to have
antitumor effects in a larger proportion of patients, and to have longer lasting effects in each patient.
One way of prolonging the effects of T cells is by immunodepletion of the regulatory immune
system, which can be achieved by total body irradiation and lymphodepleting chemotherapy2
.
Lymphodepletion before ACT can allow better proliferation of the reactive T cells by lowering the
numbers of regulatory T cells and myeloid-derived suppressor cells, enhancing innate immunity, and by
increasing cytokines42
. This lymphodepletion has improved results in mouse models of B16 melanoma43
.
Also, for stage IV melanoma response rates were 49% with 1x lymphodepletion and 72% with 3x
lymphodepletion44
. Finally, checkpoint inhibitors are proving to be a great way to assist cancer
therapy*for more information see 37, 38
.
Culturing
The practical problems of ACT are the time and resources necessary to culture and test these T-
cells for the 6-8 weeks31
, and a minimum of 2 extra weeks to incorporate any genetically engineered
changes required for TCR or CAR modification29
. This must be done over for every single patient (the
epitome of individualized medicine) as each cancer is different, presents unique markers, and
accordingly requires T-cells with unique receptors2
. This delay of at least 6-10 weeks makes this therapy
somewhat sluggish, and the weeks of cell culture requires hours of work for a researcher. Using current
methods, it would be unfeasible to run these ACT protocols for the general population in all but very
rare cancers.
Cost
All the time and resources necessary to culture these T-cells inherently makes this a costly
process. The hospital stay required to have the cells injected is also costly, as it is essentially a form of
transplant2
. It is nearly impossible to even estimate a cost seeing as the protocols change rapidly as
development of this field proceeds, and as costs are not always publicly reported in clinical trials2
. For a
very rough estimate, sipuleucel-T, a cancer vaccine used to treat prostate cancer, requires the isolation
of immune cells from each patient, in-vitro culturing, and infusion back into the patient, all just like ACT,
and it costs $93,000 all told44
.
THE FUTURE FOR ACT
ACT is certainly still a treatment under development. Its simple premise inspires hope for
success in the future, but the long list of limitations admittedly casts doubt. What will likely determine if
ACT becomes a widely used therapy in the near future is how the cost-to-benefit ratio changes in
relation to other therapies like chemotherapy and radiation. Unless the process of T-cell culture and
engineering can be majorly streamlined and automated, it seems unlikely that ACT will be able to
compete with chemotherapy and radiation therapy for most cancers.
Of course, the cost-to-benefit ratio goes hand in hand with the biological effectiveness of ACT.
Many versions of this therapy pose a toxicity risk to the patient, which is often quite severe. While this
may be quite difficult or even impossible to eliminate completely given the need for enhanced affinity to
self-antigens, there needs to be a way of minimizing the potential harm to the patient. This could come
in the form of an inducible apoptosis mechanism for the modified T cells, or by focusing efforts on
neoantigens as receptor targets rather than self-antigens. These engineered T cells must also last longer
in the host in order to produce significant anti-tumor effects, so these cells need to proliferate better
and avoid degradation. Finally, better receptors need to be engineered for use in TCR and CARs, though
given the transformation between first and third generation CARs there seems to be little doubt that
these improvements will come with time.
To its credit, ACT could eventually enable patients to attack even aggressive, metastatic, and
heterogeneous cancers with very limited toxicity or side effects. Perhaps this dream will propel the
research needed to bring this form of therapy to fruition.
References
1. Hanahan D, and Weinberg R A (2011) Hallmarks of Cancer: The Next Generation. Cell. 144:646-
674
2. Perica K, Varela JC, Oelke M, Schneck J (2015) Adoptive T Cell Immunotherapy for Cancer.
Rambam Maimonides Med J 6: 1-7
3. Schreiber RD, Old LJ, Smyth MJ (2011) Cancer immunoediting: integrating immunity's roles in
cancer suppression and promotion. Science 331:1565–70
4. Schlom J, Arlen PM, Gulley JL (2007) Cancer vaccines: moving beyond current paradigms. Clin
Cancer Res 13:3776–82
5. Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA (2004) Generation of tumor-
infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J
Immunother 26:332–42
6. Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, Kammula US, Royal RE,
Sherry RM, Wunderlich JR et al (2009) Gene therapy with human and mouse T cell receptors
mediates cancer regression and targets normal tissues expressing cognate antigen. Blood
114:535–546
7. Kalos M, June CH (2013) Adoptive T cell transfer for cancer immunotherapy in the era of
synthetic biology. Immunity 39:49–60
8. Vonderheide RH, June AH (2014) Engineering T cells for cancer: our synthetic future.
Immunological Reviews 257:7-13
9. Aleksic M, Liddy N, Molloy PE, Pumphrey N, Vuidepot A, Chang KM, Jakobsen BK (2012)
Different affinity windows for virus and cancer-specific T-cell receptors: implications for
therapeutic strategies. Eur J Immunol 42:3174–3179
10. June C, Maus M, Plesa G, et. al. (2014) Engineered T cell for cancer therapy. Cancer Immunol.
Immunother. 63:969-975
11. Jensen MC, Riddell SR (2014) Design and implementation of adoptive therapy with chimeric
antigen receptor-modified T cells. Immunol Rev 257:127–144
12. Simpson A, Caballero O, Jungbluth A, Chen Y, Old L (2005) Cancer/testis antigens, gametogenesis
and cancer. Nat Rev Cancer 5:615–625
13. Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, Wunderlich JR, Nahvi AV,
Helman LJ, Mackall CL (2011) Tumor regression in patients with metastatic synovial cell sarcoma
and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol
29:917
14. Cameron BJ, Gerry AB, Dukes J, Harper JV, Kannan V, Bianchi FC, Grand F, Brewer JE, Gupta M,
Plesa G et al (2013) Identification of a titin-derived HLA-A1-presented peptide as a crossreactive
target for engineered MAGE A3-Directed T cells. Sci Transl Med 5(197):197ra103.
doi:10.1126/scitranslmed.3006034
15. . Linette GP, Stadtmauer EA, Maus MV, Rapoport AP, Levine BL, Emery L, Litzky L, Bagg A,
Carreno BM, Cimino PJ et al (2013) Cardiovascular toxicity and titin cross-reactivity of affinity
enhanced T cells in myeloma and melanoma. Blood 122:863–871
16. Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, Stetler-Stevenson M,
Phan GQ, Hughes MS, Sherry RM et al (2012) B-cell depletion and remissions of malignancy
along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-
transduced T cells. Blood 119:2709–2720
17. Tang Y, Xu X, Song H, Yang S, Shi S, Wei J, Pan B, Zhao F, Liao C, Luo C (2008) Early diagnostic and
prognostic significance of a specific Th1/Th2 cytokine pattern in children with haemophagocytic
syndrome. Br J Haematol 143:84–91
18. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-Ojeda O,
Olszewska M et al (2014) Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell
acute lymphoblastic leukemia. Sci Transl Med 6(224):224ra225.
doi:10.1126/scitranslmed.3008226
19. Saha B, Harlan DM, Lee KP, June CH, Abe R (1996) Protection against lethal toxic shock by
targeted disruption of the CD28 gene. J Exp Med 183:2675–2680
20. . Klinger M, Brandl C, Zugmaier G, Hijazi Y, Bargou RC, Topp MS, Gokbuget N, Neumann S,
Goebeler M, Viardot A et al (2012) Immunopharmacologic response of patients with B-lineage
acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE
antibody blinatumomab. Blood 119:6226–6233
21. Irving BA, Weiss A (1991) The cytoplasmic domain of the T cell receptor zeta chain is sufficient to
couple to receptor-associated signal transduction pathways. Cell 64:891–901
22. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk
E (2006) Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes
genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol
24:e20–e22
23. . Finney HM, Lawson ADG, Bebbington CR, Weir ANC (1998) Chimeric receptors providing both
primary and costimulatory signaling in T cells from a single gene product. J Immunol 161:2791–
2797
24. Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S (2000) Escape of human solid tumors from T-cell
recognition: molecular mechanisms and functional significance. Adv Immunol 74:181–273
25. Vitale M, Pelusi G, Taroni B, Gobbi G, Micheloni C, Rezzani R, Donato F, Wang X, Ferrone S
(2005) HLA class I antigen downregulation in primary ovary carcinoma lesions: association with
disease stage. Clin Cancer Res 11:67–72
26. Scholler J, Brady T, Binder-Scholl G, Hwang W-T, Plesa G, Hege K, Vogel A, Kalos M, Riley J, Deeks
S et al (2012) Decade-long safety and function of retroviral-modified chimeric antigen receptor T
cells. Sci Transl Med 4(132):132Ra153. doi:10.1126/scitran slmed.3003761
27. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C,
Olszewska M et al (2013) CD19- targeted T cells rapidly induce molecular remissions in adults
with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 5(177):177ra138.
doi:10.1126/scitranslmed.3005930
28. Davis MM, Krogsgaard M, Huse M, Huppa J, Lillemeier BF, Li QJ (2007) T cells as a self-
referential, sensory organ. Annu Rev Immunol 25:681–695
29. Wang X, Riviere I (2015) Manufacture of tumor- and virus-specefic T lymphocytes for adoptive
cell thearpies. Cancer Gene Therapy 22:85-94
30. Drake CG, Jaffee E, Pardoll DM (2006) Mechanisms of immune evasion by tumors. Adv Immunol
90:51–81
31. Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA (2003) Generation of tumor-
infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J
Immunother 26: 332–342
32. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM et al (2006) Cancer
regression in patients after transfer of genetically engineered lymphocytes. Science 314: 126–
129
33. Hollyman D, Stefanski J, Przybylowski M, Bartido S, Borquez-Ojeda O, Taylor C et al (2009)
Manufacturing validation of biologically functional T cells targeted to CD19 antigen for
autologous adoptive cell therapy. J Immunother 32: 169–180
34. Hogquist KA, Baldwin TA, Jameson SC (2005) Central tolerance: learning self-control in the
thymus. Nat Rev Immunol 5:772–82
35. Wherry EJ (2011) T cell exhaustion. Nat Immunol 131:492–9
36. Gorelik L, Flavell RA (2001) Immune-mediated eradication of tumors through the blockade of
transforming growth factor-beta signaling in T cells. Nat Med 7:1118–22
37. Hodi FS, O'Day SJ, McDermott DF, et al (2010) Improved survival with ipilimumab in patients
with metastatic melanoma. N Engl J Med 363:711–23
38. Topalian S, Hodi SF, Brahmer JR, et al (2012) Safety, activity, and immune correlates of anti–PD-
1 antibody in cancer. N Engl J Med 366:2443–54
39. Rabinovich GA, Gabrilovich D, Sotomayor EM (2007) Immunosuppressive strategies that are
mediated by tumor cells. Annu Rev Immunol 25:267–96
40. Jena B, Dotti G, Cooper L (2010) Redirecting T-cell specificity by introducing a tumor-specific
chimeric antigen receptor. Blood 116:1035–1044
41. Porter DL, Levine BL, Kalos M, Bagg A, June CH (2011) Chimeric antigen receptor-modified T cells
in chronic lymphoid leukemia. N Engl J Med 365:725–733
42. Wrzesinski C, Paulos CM, Kaiser A, et al (2010) Increased intensity lymphodepletion enhances
tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother 33:1–7
43. Gattinoni L, Finkelstein SE, Klebanoff CA, et al (2005) Removal of homeostatic cytokine sinks by
lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J
Exp Med 202:907–12
44. Lesterhuis WJ, Haanen JB, Punt CJ (2011) Cancer immunotherapy - revisited. Nat Rev Drug
Discov 10:591–600

Contenu connexe

Tendances

Nigella sativa bioactives against Non-Small Cell Lung Cancer & Breast Cancer
Nigella sativa bioactives against Non-Small Cell Lung Cancer & Breast CancerNigella sativa bioactives against Non-Small Cell Lung Cancer & Breast Cancer
Nigella sativa bioactives against Non-Small Cell Lung Cancer & Breast CancerYusuf Asad
 
Immunotherapy Web Presentation
Immunotherapy   Web PresentationImmunotherapy   Web Presentation
Immunotherapy Web Presentationanthonymaida
 
Content Cytotoxicity Studies of Colorectal Carcinoma Cells Using Printed Impe...
Content Cytotoxicity Studies of Colorectal Carcinoma Cells Using Printed Impe...Content Cytotoxicity Studies of Colorectal Carcinoma Cells Using Printed Impe...
Content Cytotoxicity Studies of Colorectal Carcinoma Cells Using Printed Impe...journalBEEI
 
Principles of cancer immunotherapy
Principles of cancer immunotherapyPrinciples of cancer immunotherapy
Principles of cancer immunotherapyIhor Arkhypov
 
University of Missouri - Columbia, POSH Inhibitor-Based Cancer Therapy
University of Missouri - Columbia, POSH Inhibitor-Based Cancer TherapyUniversity of Missouri - Columbia, POSH Inhibitor-Based Cancer Therapy
University of Missouri - Columbia, POSH Inhibitor-Based Cancer Therapykphodel
 
Sipuleucel_T Immunotherapy for Metastatic Prostate Cancer after Failing Hormo...
Sipuleucel_T Immunotherapy for Metastatic Prostate Cancer after Failing Hormo...Sipuleucel_T Immunotherapy for Metastatic Prostate Cancer after Failing Hormo...
Sipuleucel_T Immunotherapy for Metastatic Prostate Cancer after Failing Hormo...mjavan2001
 
Global cancer immunotherapy market outlook 2020
Global cancer immunotherapy market outlook 2020Global cancer immunotherapy market outlook 2020
Global cancer immunotherapy market outlook 2020KuicK Research
 
2015 11 Inmunoterapia en nsclc
2015 11 Inmunoterapia en nsclc2015 11 Inmunoterapia en nsclc
2015 11 Inmunoterapia en nsclcMartín Lázaro
 
Animal models in developmental therapeitocs
Animal models in developmental therapeitocsAnimal models in developmental therapeitocs
Animal models in developmental therapeitocsDr.Bhavin Vadodariya
 
Cancer Immunotherapy
Cancer ImmunotherapyCancer Immunotherapy
Cancer ImmunotherapyAsokan R
 
Cancer therapy at gene level
Cancer therapy at gene levelCancer therapy at gene level
Cancer therapy at gene levelSuganyaPaulraj
 
Tumor And Immunity
Tumor And ImmunityTumor And Immunity
Tumor And Immunity軒名 林
 
Cancer therapy at protein level
Cancer therapy at protein levelCancer therapy at protein level
Cancer therapy at protein levelSuganyaPaulraj
 
CAR-T cells in Leukemia. Prof. Keiya Ozawa
CAR-T cells in Leukemia. Prof. Keiya OzawaCAR-T cells in Leukemia. Prof. Keiya Ozawa
CAR-T cells in Leukemia. Prof. Keiya Ozawaspa718
 
HadiDiVitoMarfiaNavoneCampanellaRiboniChap
HadiDiVitoMarfiaNavoneCampanellaRiboniChapHadiDiVitoMarfiaNavoneCampanellaRiboniChap
HadiDiVitoMarfiaNavoneCampanellaRiboniChapLoubna Abdel Hadi
 
Immunobiology of cancer
Immunobiology of cancerImmunobiology of cancer
Immunobiology of cancerDiana Santos
 
Journal club_NKs CXCR2 gene modified - improved migration
Journal club_NKs CXCR2 gene modified - improved migrationJournal club_NKs CXCR2 gene modified - improved migration
Journal club_NKs CXCR2 gene modified - improved migrationIhor Arkhypov
 

Tendances (20)

Nigella sativa bioactives against Non-Small Cell Lung Cancer & Breast Cancer
Nigella sativa bioactives against Non-Small Cell Lung Cancer & Breast CancerNigella sativa bioactives against Non-Small Cell Lung Cancer & Breast Cancer
Nigella sativa bioactives against Non-Small Cell Lung Cancer & Breast Cancer
 
Immunotherapy Web Presentation
Immunotherapy   Web PresentationImmunotherapy   Web Presentation
Immunotherapy Web Presentation
 
Content Cytotoxicity Studies of Colorectal Carcinoma Cells Using Printed Impe...
Content Cytotoxicity Studies of Colorectal Carcinoma Cells Using Printed Impe...Content Cytotoxicity Studies of Colorectal Carcinoma Cells Using Printed Impe...
Content Cytotoxicity Studies of Colorectal Carcinoma Cells Using Printed Impe...
 
Ijsrp p10758
Ijsrp p10758Ijsrp p10758
Ijsrp p10758
 
Principles of cancer immunotherapy
Principles of cancer immunotherapyPrinciples of cancer immunotherapy
Principles of cancer immunotherapy
 
University of Missouri - Columbia, POSH Inhibitor-Based Cancer Therapy
University of Missouri - Columbia, POSH Inhibitor-Based Cancer TherapyUniversity of Missouri - Columbia, POSH Inhibitor-Based Cancer Therapy
University of Missouri - Columbia, POSH Inhibitor-Based Cancer Therapy
 
Sipuleucel_T Immunotherapy for Metastatic Prostate Cancer after Failing Hormo...
Sipuleucel_T Immunotherapy for Metastatic Prostate Cancer after Failing Hormo...Sipuleucel_T Immunotherapy for Metastatic Prostate Cancer after Failing Hormo...
Sipuleucel_T Immunotherapy for Metastatic Prostate Cancer after Failing Hormo...
 
Global cancer immunotherapy market outlook 2020
Global cancer immunotherapy market outlook 2020Global cancer immunotherapy market outlook 2020
Global cancer immunotherapy market outlook 2020
 
2015 11 Inmunoterapia en nsclc
2015 11 Inmunoterapia en nsclc2015 11 Inmunoterapia en nsclc
2015 11 Inmunoterapia en nsclc
 
Animal models in developmental therapeitocs
Animal models in developmental therapeitocsAnimal models in developmental therapeitocs
Animal models in developmental therapeitocs
 
Cancer Immunotherapy
Cancer ImmunotherapyCancer Immunotherapy
Cancer Immunotherapy
 
Cancer therapy at gene level
Cancer therapy at gene levelCancer therapy at gene level
Cancer therapy at gene level
 
Car T-cell therapy
Car T-cell therapyCar T-cell therapy
Car T-cell therapy
 
Tumor And Immunity
Tumor And ImmunityTumor And Immunity
Tumor And Immunity
 
New Frontiers in Precision Immuno-Oncology: Leveraging Biomarkers to Refine a...
New Frontiers in Precision Immuno-Oncology: Leveraging Biomarkers to Refine a...New Frontiers in Precision Immuno-Oncology: Leveraging Biomarkers to Refine a...
New Frontiers in Precision Immuno-Oncology: Leveraging Biomarkers to Refine a...
 
Cancer therapy at protein level
Cancer therapy at protein levelCancer therapy at protein level
Cancer therapy at protein level
 
CAR-T cells in Leukemia. Prof. Keiya Ozawa
CAR-T cells in Leukemia. Prof. Keiya OzawaCAR-T cells in Leukemia. Prof. Keiya Ozawa
CAR-T cells in Leukemia. Prof. Keiya Ozawa
 
HadiDiVitoMarfiaNavoneCampanellaRiboniChap
HadiDiVitoMarfiaNavoneCampanellaRiboniChapHadiDiVitoMarfiaNavoneCampanellaRiboniChap
HadiDiVitoMarfiaNavoneCampanellaRiboniChap
 
Immunobiology of cancer
Immunobiology of cancerImmunobiology of cancer
Immunobiology of cancer
 
Journal club_NKs CXCR2 gene modified - improved migration
Journal club_NKs CXCR2 gene modified - improved migrationJournal club_NKs CXCR2 gene modified - improved migration
Journal club_NKs CXCR2 gene modified - improved migration
 

Similaire à Review of Adoptive T-Cell Immunotherapy

Chimeric Antigen Receptors (paper with corresponding power point)
Chimeric Antigen Receptors (paper with corresponding power point)Chimeric Antigen Receptors (paper with corresponding power point)
Chimeric Antigen Receptors (paper with corresponding power point)Kevin B Hugins
 
Engineered T Cell Therapy for Gynecologic Malignancies Challenges and Opportu...
Engineered T Cell Therapy for
Gynecologic Malignancies
Challenges and Opportu...Engineered T Cell Therapy for
Gynecologic Malignancies
Challenges and Opportu...
Engineered T Cell Therapy for Gynecologic Malignancies Challenges and Opportu...RudrikaChandra1
 
Applying dosing schedules to the clinical protocols of combinatorial therapy
Applying dosing schedules to the clinical protocols of combinatorial therapyApplying dosing schedules to the clinical protocols of combinatorial therapy
Applying dosing schedules to the clinical protocols of combinatorial therapyRetired from EASTMAN KODAK
 
Vital Signs Edition #6
Vital Signs   Edition #6Vital Signs   Edition #6
Vital Signs Edition #6ScottJordan
 
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...Peter Pachmann
 
CAR-T CELLS IN SOLID TUMORS
CAR-T CELLS IN SOLID TUMORSCAR-T CELLS IN SOLID TUMORS
CAR-T CELLS IN SOLID TUMORSClaire Roudot
 
Gene therapy for the treatmen of cancer
Gene therapy for the treatmen of cancerGene therapy for the treatmen of cancer
Gene therapy for the treatmen of cancergrachea aeryndhien
 
Immunotherapy and gene therapy
Immunotherapy and gene therapyImmunotherapy and gene therapy
Immunotherapy and gene therapyashish gupta
 
Chemotherapy Friends or Foe to Cancer Immunotherapy by Prof. Mohamed L. Salem
Chemotherapy Friends or Foe to Cancer Immunotherapy by Prof. Mohamed L. SalemChemotherapy Friends or Foe to Cancer Immunotherapy by Prof. Mohamed L. Salem
Chemotherapy Friends or Foe to Cancer Immunotherapy by Prof. Mohamed L. SalemProf. Mohamed Labib Salem
 
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...CrimsonGastroenterology
 
Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics
Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer TherapeuticsCytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics
Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer TherapeuticsSachin K. S. Chauhan
 
WP_ClinDev-Immunotherapy_d03-final edits
WP_ClinDev-Immunotherapy_d03-final editsWP_ClinDev-Immunotherapy_d03-final edits
WP_ClinDev-Immunotherapy_d03-final editsChris Learn, Ph.D, PMP
 

Similaire à Review of Adoptive T-Cell Immunotherapy (20)

Chimeric Antigen Receptors (paper with corresponding power point)
Chimeric Antigen Receptors (paper with corresponding power point)Chimeric Antigen Receptors (paper with corresponding power point)
Chimeric Antigen Receptors (paper with corresponding power point)
 
Engineered T Cell Therapy for Gynecologic Malignancies Challenges and Opportu...
Engineered T Cell Therapy for
Gynecologic Malignancies
Challenges and Opportu...Engineered T Cell Therapy for
Gynecologic Malignancies
Challenges and Opportu...
Engineered T Cell Therapy for Gynecologic Malignancies Challenges and Opportu...
 
Applying dosing schedules to the clinical protocols of combinatorial therapy
Applying dosing schedules to the clinical protocols of combinatorial therapyApplying dosing schedules to the clinical protocols of combinatorial therapy
Applying dosing schedules to the clinical protocols of combinatorial therapy
 
Vital Signs Edition #6
Vital Signs   Edition #6Vital Signs   Edition #6
Vital Signs Edition #6
 
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
 
CAR-T CELLS IN SOLID TUMORS
CAR-T CELLS IN SOLID TUMORSCAR-T CELLS IN SOLID TUMORS
CAR-T CELLS IN SOLID TUMORS
 
Gene therapy for the treatmen of cancer
Gene therapy for the treatmen of cancerGene therapy for the treatmen of cancer
Gene therapy for the treatmen of cancer
 
Immunotherapy and gene therapy
Immunotherapy and gene therapyImmunotherapy and gene therapy
Immunotherapy and gene therapy
 
Proposal
ProposalProposal
Proposal
 
What is car t pipeline
What is car t pipelineWhat is car t pipeline
What is car t pipeline
 
Chemotherapy Friends or Foe to Cancer Immunotherapy by Prof. Mohamed L. Salem
Chemotherapy Friends or Foe to Cancer Immunotherapy by Prof. Mohamed L. SalemChemotherapy Friends or Foe to Cancer Immunotherapy by Prof. Mohamed L. Salem
Chemotherapy Friends or Foe to Cancer Immunotherapy by Prof. Mohamed L. Salem
 
The magic bullet theory for melanoma
The magic bullet theory for melanomaThe magic bullet theory for melanoma
The magic bullet theory for melanoma
 
Pham2018
Pham2018Pham2018
Pham2018
 
How to Integrate Perioperative Immunotherapy Into Multimodal Treatment Plans ...
How to Integrate Perioperative Immunotherapy Into Multimodal Treatment Plans ...How to Integrate Perioperative Immunotherapy Into Multimodal Treatment Plans ...
How to Integrate Perioperative Immunotherapy Into Multimodal Treatment Plans ...
 
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...
 
Parsing the Practicalities of Pathologic Response Assessment After Neoadjuvan...
Parsing the Practicalities of Pathologic Response Assessment After Neoadjuvan...Parsing the Practicalities of Pathologic Response Assessment After Neoadjuvan...
Parsing the Practicalities of Pathologic Response Assessment After Neoadjuvan...
 
Car t cell tumor board
Car  t cell tumor boardCar  t cell tumor board
Car t cell tumor board
 
Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics
Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer TherapeuticsCytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics
Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics
 
Immuno oncology wp
Immuno oncology wpImmuno oncology wp
Immuno oncology wp
 
WP_ClinDev-Immunotherapy_d03-final edits
WP_ClinDev-Immunotherapy_d03-final editsWP_ClinDev-Immunotherapy_d03-final edits
WP_ClinDev-Immunotherapy_d03-final edits
 

Review of Adoptive T-Cell Immunotherapy

  • 1. Luke Brennan 4/21/2015 Review of Adoptive T-Cell Immunotherapy INTRODUCTION One of the ‘hallmarks’ of cancer is its ability to evade detection and destruction by the body’s immune system by essentially hiding in plain sight1 . Cancer is also inherently derived from the body’s own tissue, and harbors many of the signals that identify it as ‘self,’ which contributes to its camoflauge1 . These two factors have made cancer so difficult to treat, because the immune system is insufficient and man-made chemicals and radiation therapy used to attack cancer inevitably targets unintended healthy tissue. These issues and more prompted the development of immunotherapy for cancer treatment. Much like how a vaccination conditions the adaptive immune system to respond to a certain pathogen, immunotherapy would allow the immune system to sense and destroy cancer, though it uses different means. This is often referred to as the cancer immunosurveillance hypothesis, and it gained significant support after the verification of human tumor-associated antigens (TAAs) which are identifiers unique to cancer, and can be exploited to discern cancer from healthy tissue2 . Although immunotherapy is used to treat cancer in many ways, the scope of this paper will be limited to Adoptive T-Cell Therapy (ACT). T-cells are lymphocytes, or white blood cells, which are present throughout the body and attack pathogens and cells marked for apoptosis. T cells usually destroy cells that would become cancerous before they even become a problem, when these mutated cells aren’t destroyed and proliferate in an uncontrolled way they become cancers. ACT is the process by which T- cells (originally extracted from the body, in most cases and for the purposes of this paper) with cancer- recognizing properties are introduced into the patient to combat cancer. Harnessing the power of these cancer sensitive T cells for use against cancer has great potential for many reasons: “1) T cell responses are specific, and can thus potentially distinguish between healthy and cancerous tissue; 2) T cells responses are robust, undergoing up to 1,000-fold clonal expansion after activation; 3) T cell response can traffic to the site of antigen, suggesting a mechanism for eradication of distant metastases; 4) T cell responses have memory, maintaining therapeutic effect for many years after initial treatment.”2 As elegant a solution as immunotherapy may seem for the above reasons, many challenges must be overcome before this becomes a viable treatment for mainstream cancer patients, as discussed later in this review. Regardless, the following sections describe two main types of ACT based on the anti- tumor activity of the lymphocytes collected from the patient. ACT USING TUMOR-INFILTRATING LYMPHOCYTES This type of ACT is dependent on the collection of lymphocytes with anti-tumor cytotoxic activity, called tumor-infiltrating lymphocytes (TILs), against the cancer in question2 . Fortunately, such T- cells have been identified in the tumor samples of up to 80% of melanoma patients, with admittedly lower frequency in other forms of cancer5 . In fact, though not universal, lymphocyte infiltration of tumor tissue has in fact become a hallmark of cancer1 . In the cases where such cells are detected and can be extracted, these TILs can be cultured and grown ex vivo.
  • 2. This process begins with the TILs from a “micro culture derived from a single tumor fragment or 106 viable cells derived from a single-cell enzymatic digestion [of a resected tumor specimen]”29 . These cells are grown and cultures split as needed for two weeks with high dose interleukin 2, after which point each culture is kept at 0.8-1.6x106 mL-1 in flasks and generates roughly 5x107 cells (the required minimum being 3x107 cells) from each culture after 3-5 weeks29 . The cultures are then tested for activity and specificity by an immunosorbent assay after stimulation with tumor cells, and cultures deemed active undergo a rapid expansion phase for 2 weeks with donor feeder cells, anti-CD3 OKT-3 monoclonal antibody, and high dose interleukin 2 to promote expansion 29 . In total, the entire manufacture process takes roughly 6-8 weeks31 . The expanded colonies of TILs are then reintroduced to the body to attack the cancer with greater numbers than before. This approach capitalizes on the ‘strength in numbers’ strategy, but with the added benefit of more specific TILs from the testing and selection processes performed ex-vivo29 . This type of ACT would be considered the most feasible, but it sacrifices efficacy. ACTs using TILs are limited in a few ways. First, this treatment is simply not an option for any patient in whom TILs cannot be identified or extracted. Even for those patients from whom TILs can be extracted, there is growing concern that these TILs may have little to no effect on tumor progression despite their presence in areas of tumor inhibition2 . Further, to avoid unintended autoimmune attack, these TILs generally have low affinity to the self-antigens and germline antigens that are usually overexpressed by cancerous tissues (but are shared by healthy body tissues)6 . While this is still the most effective method of ACT to date, these limitations have encouraged development of the second type of ACT. ACT USING ENGINEERED T CELLS This method of ACT makes use of the T-cells’ time ex vivo (as described in the methods section) to enhance T-cell function by changing receptors to give the cells improved affinity to self-antigens or novel specificity, and/or by improving their proliferative capacity through alterations in signaling functions7 . Because the cells are being engineered, the T-cells do not necessarily need to be TILs or even from the tumor region, and can be collected from peripheral blood samples which are much easier to collect, and are present in every patient8 . This higher affinity and novel specificity is largely achieved through expression of heterodimeric T-cell receptors (TCRs) in T-cells which can allow tighter binding to the self-antigens overexpressed on cancer, or neo-antigens, which are epitopes that are only present in cells with cancerous mutations9 . To achieve this, a gene corresponding to the engineered TCR is introduced into the T cells by a retroviral vector32 . The TCR then “combines TCR-alpha and TCR-beta genes” and undergoes MHC restriction, a process that ensures it only recognizes peptide antigens when they are presented on the body’s MHC molecule10 . Another method to enhance affinity is to reduce N- glycosylation on TCR chains11 . Surprisingly, self-antigens (specifically genes of the cancer testis family*for more info look at 12 ) rather than neoantigens, are more often the targets of such engineered TCRs12 . Some of the first trials of this type have been for the NY ESO-1-specific and HLA-A2-restricted TCR for use in melanoma, myeloma, and synovial cell sarcoma13 . These studies have been making great progress and have not shown toxicity. Toxicity is a large concern in these trials, after all these cells are being engineered to attack self-antigens that are present on healthy body tissue. In fact in another study a TCR engineered for enhanced MAGE 3 affinity and HLA-A1-restriction had off target recognition of the muscle protein Titin, resulting in lethal cardiac toxicity for both patients14,15 . This will be discussed further along in the paper. Another problem of ACT with genetically engineered T-cells is that, due to a multitude of factors known and unknown, it has been less successful than ACT with TILs16,17 . This being said, ACT remains relevant because of how much it can improve with better protocols, and because it
  • 3. greatly expands the range of ACT to cancers like neuroblastoma, synovial cell carcinoma, and colorectal carcinoma (among others) where TILs cannot be extracted18,19, 20 . The synthesis of these genetically engineered T-cells begins with T-cells from ficoll-purified PMBCs, which are activated with OKT-3 antibodies29 . They are then transduced with a retroviral vector that encodes the desired antigen-specific TCR, cultured for 2 weeks, and transduced and expanded under cGMP 32,33 . These activated T-cells are then transduced again with retroviral vectors in RetroNectin-coated cell bags, inoculated in a WAVE bioreactor for two days, and expanded with a continuous perfusion regime29 . After this, the beads (from cGMP treatment) are removed and the cells are formulated for infusion29 . This total process takes about 2 weeks and is of a large enough scale to support multiple clinical trials29 . CAR T-Cells One other noteworthy category of genetically engineered T-cells for use in ACT is the CAR- modified T-cell. This modification exploits the fact that “the cytoplasmic tail of the TCR CD3zeta chain [can] activate T-cells without the rest of the receptor complex”21 . Despite disappointing results from first generation CARs22 , second and third generation CARs have shown antitumor effects and have remained in patients for up to a decade (but also as little as a month)*for more information about the modifications of second and third CARs go to 23 . What is promising is that response rates are generally encouraging, over 50% of patients achieving some kind of remission in studies at the University of Pennsylvania, and that this response is correlated with CAR T-cell proliferation, indicating that the remission is related to the presence or activities of the CAR T-cells10 . CAR modification represent a valuable alternative to TCR modification because only CAR- modified T-cells can recognize a cancer cell that has lost MHC expression, a condition which applies to many cancers24,25 . Also, CAR-modified T-cells are sometimes able to remain in the body and fight cancer much longer than TCR-modified T-cells, though the longevity of each varies substantally26,27 . This being said, TCR-modification has its place, as it is “able to sense the entire intracellular proteome that is presented by MHC molecules” and accordingly can attack a wider range of molecules in the cancer cells10 . It also requires about 10x less target antigen expression (sometimes less than 10 ligands) to induce cytolosys28 . DISCUSSION ACT involving any type of T-cell is not yet ready for the standard treatment of cancers on a large scale. This is because of several factors both biological and practical. T-cell Inhibition First, both the cancer and the immune system itself have several methods of inhibiting an immune response to cancer tissue. As T-cells are maturing in the thymus, they are tested for recognition to self-antigens, and those that are particularly reactive are deleted to avoid autoimmunity34 . These especially active T-cells that the body stops from maturing are needed to recognize the over-expressed self-antigens on the surface of cancer cells, so this limits the effectiveness of any TIL related ACT10 . T- cells are also inhibited from attacking cancer in maturity by peripheral tolerance mechanisms, which consists of 3 categories2 : 1. Inhibition from the T-cell itself or its nature, like low activity around inflammation*for more information see 35
  • 4. 2. Inhibition from the Tumor, like the expression of checkpoint molecules*for more information go to 36, this is an area of relatively high development after trials in melanoma (37,38). 3. Inhibition from regulatory T-cells and myeloid derived suppressor cells*for more information see 39 . The first two sources of inhibition are difficult to handle, as they are meant to stop autoimmune attack on one’s own body tissues2 . Simply turning these inhibitory mechanisms off could cause severe autoimmune disease. Toxicity Toxicity is another huge concern, with off-target autoimmunity having lethal ramifications in both TCR-modified T-cell ACT studies (see above) and CAR-modified T-cells which can cause B-cell aplasia, lysis syndrome, and cytokine release syndrome22,40,41,10 . To make matters works, the toxicity from these ACTs can target the brain or heart, and cannot always be managed with corticosteroids10 . The problem here is that T-cells are being engineered to have greater recognition of self-antigens, and while this allows them to detect the overexpressed self-antigens on cancer cells, it is difficult to eliminate all off target effects since almost every cell in the body has these self-antigens. It goes without saying that work is being done to stop this off target autoimmunity, and one noteworthy idea is to add inducible mechanisms for T-cell apoptosis when engineering the T-cells ex-vivo so that they could be stopped quickly and easily if toxicity were to become an issue10 . Longevity The final biological issue with ACT is response and longevity, specifically for engineered T-cells. As noted above, most TCR-modified T-cells can often last for only a few months, and CAR-modified T- cells aren’t much better. This is only counting the patients who respond well to the treatment. For ACT to become a mainstream treatment, researchers need to find ways to allow the engineered cells to have antitumor effects in a larger proportion of patients, and to have longer lasting effects in each patient. One way of prolonging the effects of T cells is by immunodepletion of the regulatory immune system, which can be achieved by total body irradiation and lymphodepleting chemotherapy2 . Lymphodepletion before ACT can allow better proliferation of the reactive T cells by lowering the numbers of regulatory T cells and myeloid-derived suppressor cells, enhancing innate immunity, and by increasing cytokines42 . This lymphodepletion has improved results in mouse models of B16 melanoma43 . Also, for stage IV melanoma response rates were 49% with 1x lymphodepletion and 72% with 3x lymphodepletion44 . Finally, checkpoint inhibitors are proving to be a great way to assist cancer therapy*for more information see 37, 38 . Culturing The practical problems of ACT are the time and resources necessary to culture and test these T- cells for the 6-8 weeks31 , and a minimum of 2 extra weeks to incorporate any genetically engineered changes required for TCR or CAR modification29 . This must be done over for every single patient (the epitome of individualized medicine) as each cancer is different, presents unique markers, and accordingly requires T-cells with unique receptors2 . This delay of at least 6-10 weeks makes this therapy somewhat sluggish, and the weeks of cell culture requires hours of work for a researcher. Using current methods, it would be unfeasible to run these ACT protocols for the general population in all but very rare cancers. Cost
  • 5. All the time and resources necessary to culture these T-cells inherently makes this a costly process. The hospital stay required to have the cells injected is also costly, as it is essentially a form of transplant2 . It is nearly impossible to even estimate a cost seeing as the protocols change rapidly as development of this field proceeds, and as costs are not always publicly reported in clinical trials2 . For a very rough estimate, sipuleucel-T, a cancer vaccine used to treat prostate cancer, requires the isolation of immune cells from each patient, in-vitro culturing, and infusion back into the patient, all just like ACT, and it costs $93,000 all told44 . THE FUTURE FOR ACT ACT is certainly still a treatment under development. Its simple premise inspires hope for success in the future, but the long list of limitations admittedly casts doubt. What will likely determine if ACT becomes a widely used therapy in the near future is how the cost-to-benefit ratio changes in relation to other therapies like chemotherapy and radiation. Unless the process of T-cell culture and engineering can be majorly streamlined and automated, it seems unlikely that ACT will be able to compete with chemotherapy and radiation therapy for most cancers. Of course, the cost-to-benefit ratio goes hand in hand with the biological effectiveness of ACT. Many versions of this therapy pose a toxicity risk to the patient, which is often quite severe. While this may be quite difficult or even impossible to eliminate completely given the need for enhanced affinity to self-antigens, there needs to be a way of minimizing the potential harm to the patient. This could come in the form of an inducible apoptosis mechanism for the modified T cells, or by focusing efforts on neoantigens as receptor targets rather than self-antigens. These engineered T cells must also last longer in the host in order to produce significant anti-tumor effects, so these cells need to proliferate better and avoid degradation. Finally, better receptors need to be engineered for use in TCR and CARs, though given the transformation between first and third generation CARs there seems to be little doubt that these improvements will come with time. To its credit, ACT could eventually enable patients to attack even aggressive, metastatic, and heterogeneous cancers with very limited toxicity or side effects. Perhaps this dream will propel the research needed to bring this form of therapy to fruition. References 1. Hanahan D, and Weinberg R A (2011) Hallmarks of Cancer: The Next Generation. Cell. 144:646- 674 2. Perica K, Varela JC, Oelke M, Schneck J (2015) Adoptive T Cell Immunotherapy for Cancer. Rambam Maimonides Med J 6: 1-7 3. Schreiber RD, Old LJ, Smyth MJ (2011) Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 331:1565–70 4. Schlom J, Arlen PM, Gulley JL (2007) Cancer vaccines: moving beyond current paradigms. Clin Cancer Res 13:3776–82 5. Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA (2004) Generation of tumor- infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J Immunother 26:332–42 6. Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, Kammula US, Royal RE, Sherry RM, Wunderlich JR et al (2009) Gene therapy with human and mouse T cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 114:535–546
  • 6. 7. Kalos M, June CH (2013) Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity 39:49–60 8. Vonderheide RH, June AH (2014) Engineering T cells for cancer: our synthetic future. Immunological Reviews 257:7-13 9. Aleksic M, Liddy N, Molloy PE, Pumphrey N, Vuidepot A, Chang KM, Jakobsen BK (2012) Different affinity windows for virus and cancer-specific T-cell receptors: implications for therapeutic strategies. Eur J Immunol 42:3174–3179 10. June C, Maus M, Plesa G, et. al. (2014) Engineered T cell for cancer therapy. Cancer Immunol. Immunother. 63:969-975 11. Jensen MC, Riddell SR (2014) Design and implementation of adoptive therapy with chimeric antigen receptor-modified T cells. Immunol Rev 257:127–144 12. Simpson A, Caballero O, Jungbluth A, Chen Y, Old L (2005) Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer 5:615–625 13. Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, Wunderlich JR, Nahvi AV, Helman LJ, Mackall CL (2011) Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol 29:917 14. Cameron BJ, Gerry AB, Dukes J, Harper JV, Kannan V, Bianchi FC, Grand F, Brewer JE, Gupta M, Plesa G et al (2013) Identification of a titin-derived HLA-A1-presented peptide as a crossreactive target for engineered MAGE A3-Directed T cells. Sci Transl Med 5(197):197ra103. doi:10.1126/scitranslmed.3006034 15. . Linette GP, Stadtmauer EA, Maus MV, Rapoport AP, Levine BL, Emery L, Litzky L, Bagg A, Carreno BM, Cimino PJ et al (2013) Cardiovascular toxicity and titin cross-reactivity of affinity enhanced T cells in myeloma and melanoma. Blood 122:863–871 16. Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, Stetler-Stevenson M, Phan GQ, Hughes MS, Sherry RM et al (2012) B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor- transduced T cells. Blood 119:2709–2720 17. Tang Y, Xu X, Song H, Yang S, Shi S, Wei J, Pan B, Zhao F, Liao C, Luo C (2008) Early diagnostic and prognostic significance of a specific Th1/Th2 cytokine pattern in children with haemophagocytic syndrome. Br J Haematol 143:84–91 18. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-Ojeda O, Olszewska M et al (2014) Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 6(224):224ra225. doi:10.1126/scitranslmed.3008226 19. Saha B, Harlan DM, Lee KP, June CH, Abe R (1996) Protection against lethal toxic shock by targeted disruption of the CD28 gene. J Exp Med 183:2675–2680 20. . Klinger M, Brandl C, Zugmaier G, Hijazi Y, Bargou RC, Topp MS, Gokbuget N, Neumann S, Goebeler M, Viardot A et al (2012) Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood 119:6226–6233 21. Irving BA, Weiss A (1991) The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor-associated signal transduction pathways. Cell 64:891–901 22. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E (2006) Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol 24:e20–e22
  • 7. 23. . Finney HM, Lawson ADG, Bebbington CR, Weir ANC (1998) Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol 161:2791– 2797 24. Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S (2000) Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol 74:181–273 25. Vitale M, Pelusi G, Taroni B, Gobbi G, Micheloni C, Rezzani R, Donato F, Wang X, Ferrone S (2005) HLA class I antigen downregulation in primary ovary carcinoma lesions: association with disease stage. Clin Cancer Res 11:67–72 26. Scholler J, Brady T, Binder-Scholl G, Hwang W-T, Plesa G, Hege K, Vogel A, Kalos M, Riley J, Deeks S et al (2012) Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med 4(132):132Ra153. doi:10.1126/scitran slmed.3003761 27. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C, Olszewska M et al (2013) CD19- targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 5(177):177ra138. doi:10.1126/scitranslmed.3005930 28. Davis MM, Krogsgaard M, Huse M, Huppa J, Lillemeier BF, Li QJ (2007) T cells as a self- referential, sensory organ. Annu Rev Immunol 25:681–695 29. Wang X, Riviere I (2015) Manufacture of tumor- and virus-specefic T lymphocytes for adoptive cell thearpies. Cancer Gene Therapy 22:85-94 30. Drake CG, Jaffee E, Pardoll DM (2006) Mechanisms of immune evasion by tumors. Adv Immunol 90:51–81 31. Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA (2003) Generation of tumor- infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J Immunother 26: 332–342 32. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM et al (2006) Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314: 126– 129 33. Hollyman D, Stefanski J, Przybylowski M, Bartido S, Borquez-Ojeda O, Taylor C et al (2009) Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother 32: 169–180 34. Hogquist KA, Baldwin TA, Jameson SC (2005) Central tolerance: learning self-control in the thymus. Nat Rev Immunol 5:772–82 35. Wherry EJ (2011) T cell exhaustion. Nat Immunol 131:492–9 36. Gorelik L, Flavell RA (2001) Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat Med 7:1118–22 37. Hodi FS, O'Day SJ, McDermott DF, et al (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363:711–23 38. Topalian S, Hodi SF, Brahmer JR, et al (2012) Safety, activity, and immune correlates of anti–PD- 1 antibody in cancer. N Engl J Med 366:2443–54 39. Rabinovich GA, Gabrilovich D, Sotomayor EM (2007) Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 25:267–96 40. Jena B, Dotti G, Cooper L (2010) Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood 116:1035–1044 41. Porter DL, Levine BL, Kalos M, Bagg A, June CH (2011) Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 365:725–733 42. Wrzesinski C, Paulos CM, Kaiser A, et al (2010) Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother 33:1–7
  • 8. 43. Gattinoni L, Finkelstein SE, Klebanoff CA, et al (2005) Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 202:907–12 44. Lesterhuis WJ, Haanen JB, Punt CJ (2011) Cancer immunotherapy - revisited. Nat Rev Drug Discov 10:591–600