Immune Response Against Tumors, Cancer, Antigens and Immunotherapy Flashcards

1
Q

Cancer

A
  • Cancer represents uncontrolled growth of transformed cells of the organism.
  • Cancer presents a challenge for the immune system as cancer arises from our normal cells to which our immune system is tolerant, but the transformation process potentially produces new determinants that may be immunogenic.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Immungenic Tumors

A
  • Immunogenic tumors are (should be) rejected due to expression of antigens that are recognized by the immune system.
  • Tumors are (should be) “seen” by T cells (both CD4+ and CD8+) using TCRs in the same fashion as T cells “see” antigen presented by APCs.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Types of Tumor Antigens

A
  1. Tumor-specific antigens (TSA)
  2. Tumorassociated antigens (TAA)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Tumor-Specific Antigens - expressed UNIQUELY in tumor cells

A
  • Viral proteins (e.g., env, gag, EBNA 3)
  • Translocations or point mutations

– BCR: ABL translocation encodes new peptides that the immune system has not been tolerized against.

-Activating mutations in Ras, or inactivating mutations in p53 are examples of point mutations.

  • UV induced tumors, leading to mutations in normal host proteins (e.g., HSPgp96 -UV sarcoma)
  • Chemically induced tumors (e.g., Heat shock proteins-Hsp70, Hsp90, gp 96)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Tumor-Associated Ags - Normal host proteins expressed in the wrong place

A

• Cancer/testis antigen (CT Ags)

-These proteins are normally expressed only in testis and not expressed on normal somatic cells including melanocytes; however, cancer cells of different types (breast, lung, ovarian, etc.) overexpress these proteins and can serve as targets for immune attack.

•Prostate specific antigen (PSA)

  • increased plasma levels in prostatic adenocarcinoma.
  • Modestly elevated in patients with benign prostatic hypertrophy.

•Mucins (MUC1, MUC4)

  • associated with breast, colon and pancreatic cancers.
  • Normal and malignant epithelial cells produce mucin. In cancer, due to under-glycosylation, the cryptic epitopes in the protein core are exposed and recognized by cytotoxic T lymphocytes.

•HER-2/neu

  • transmembrane protein that is related to platelet-derived growth factor (PDGF) receptor.
  • Over-expressed in 20% of human breast cancers and is associated with aggressive disease

•Oncofetal Ag: present during embryonic/fetal life, then diminish and reappear in malignancies

  • Carcinoembryonic antigen (CEA), a glycoprotein found in alimentary tract, pancreas, liver between 2-6 months of gestation, present at low levels in colon, lung and breast cancers. Not useful for detecting early cancer but blood levels can be used to monitor the efficacy of therapy.
  • Alpha fetoprotein (AFP) - produced primarily by fetal liver and yolk sac, exists in small quantities in normal adult serum. Elevated in some patients with liver cancer and non-malignant liver disease (cirrhosis).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

However, despite the presence of immunologically recognized antigens in many instances the cellular immune system cannot defeat the cancer due to one or more of the following factors:

A
  • Insufficient numbers and/or decreased avidity of the anti-tumor T cells due to selftolerization mechanisms, i.e., the frequency of tumor antigen-specific T cells is too low to be clinically effective.
  • *Tumor cells can be ineffective antigen-presenting cells, due to lack of proper cytokines and/or secondary signals (such as absence of B7 which may induce anergy).
  • Poor T cell trafficking to tumor site due to lack of sufficient vasculature
  • *Active suppression of immune response by regulatory/suppressor lymphocytes (Treg cells) or suppressor myeloid populations
  • *Down-regulation of MHC and peptide processing molecules that can “hide” tumors from CTL recognition and lysis.
  • *Tumors can secrete suppressive cytokines such as TGFb which stimulates accumulation of T regulatory cells (Treg) and dampens the immune response.
  • *T cell exhaustion due to the actions of CTLA-4 and PD-1 molecules.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Strategies to increase the effectiveness of cancer immunotherapy:

A
  • *Lymphodepletion of regulatory (suppressor) T cells accompanied by adoptive transfer of cytotoxic T lymphocytes (by radiation or drugs such as Cytoxan)
  • *Lymphodepletion to create a partially lymphopenic environment, creating “space” for expansion of anti-tumor T cells
  • Patient-specific therapy as practiced in CAR-T therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Four General Strategies for more effective cancer immunotherapy:

A
  1. Cancer vaccines – to generate/augment anti-tumor response:
  • Innate/nonspecific immunity – e.g., via BCG, C. parvum, cytokines (IL-2, IFN-I) injections
  • Adaptive immunity – e.g., via tumor peptide, protein, RNA, DNA, tumor lysate, tumor extract (heat-shock proteins), or “loaded” DC injections
  • Results have been marginal and mixed for both approaches
  1. Remove immune suppression and enhance anti-tumor immune response (e.g., Treg depletion, CTLA-4 blockade alone, or in conjunction with cancer vaccines or with blockade of other checkpoint inhibitory receptors such as PD-L1, etc.). Objective results quite promising!
  2. Adoptive cellular therapies –e.g., LAK (lymphokine-activated killer) cells, peptide-expanded or non-specifically stimulated anti-tumor T cells, and CAR-T with excellent outcomes
  3. Antibody-based anti-cancer therapies - the use of monoclonal Ab against cancer cell surface markers is extremely effective in some cancers.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Non-Specific Cancer Immunotherapy - Cytokines

A
  • Interleukin-2 nonspecifically stimulates proliferation and activation of T cells and NK cells. Effective in treatment of melanoma and renal cancer (~10% objective clinical responses).
  • Type I Interferons such as Interferon-alpha has been extensively used in clinical trials. Effective in treatment of numerous cancers including hairy cell leukemia, chronic myelogenous leukemia, lymphomas, AIDS-associated Kaposi’s sarcoma, and melanoma. Likely operates by killing tumor cells (particularly myeloid) directly as well as providing and/or inducing signals 2/3 (i.e., IL-12 &18 for IFN-I)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Non-Specific Cancer Immunotherapy - Bacteria and Bacterial Products

A
  1. BCG (Baceille-Calmette-Guerin)
  • Intravesicular instillation of BCG is effective in treatment of superficial bladder cancer.
  • Also used as an adjuvant to boost the effectiveness of cancer vaccines
  1. Microbial products

• Developed to avoid risks associated with use of intact organisms. Cell wall skeleton of BCG, Lipid A, and Monophosphoryl lipid A (MPL) have been used.

*Both work via binding to TLRs and other innate sensors to boost immunity and/or inflammatory response

*(Mechanisms poorly understood)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Tumor Specific Vaccines

A

•Target viral proteins - If oncogenic viruses are involved in tumorigenesis, vaccination against the virus will also be an effective anti-cancer measure.

  • Ceravix and Gardasil vaccines for papilloma virus and cervical cancer
  • HBV vaccine and hepatocellular carcinoma.
  • Peptide antigen vaccines: mutated Ras, mutated p53, Her2/neu, MART-1, gp100, MUC1, and CEA antigens are being used to vaccinate cancer patients.
  • Dendritic cells pulsed with peptides/tumor lysates, DC-tumor cell hybrid vaccines (numerous clinical trials) -Sipuleucel-T for prostate cancer.

*To date, no cytokine, non-specific or tumor vaccine has displayed significant clinical efficacy, other than the HPV vaccine vs. cervical cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Immunomodulation of Immune Suppression

A
  • A recent development in the field of cancer immunotherapy is the use of checkpoint blockades. That is, using antibodies to release the negative regulators of immune activation (the immune checkpoints).
  • This goal has been accomplished by using mAbs directed against CTLA-4 and the PD-1 pathway, alone or in combination. [NOBEL PRIZE 2018!!!]

*Remember that CTLA-4 interacts with CD80/CD86 to inhibit CD28-mediated T cell activation. Antibodies against CTLA-4 prevent this inhibition. PD-1 is the programmed cell death receptor that interacts with PD-L1 to trigger apoptosis in T cells. Antibodies against either PD-1 or PD- L1 can prevent this event from occurring.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Immunomodulation of Immune Suppression - Keytruda

A
  • Immune Checkpoint Blockade
  • There are currently multiple FDA-approved antibodies for this application; Keytruda was the first one approved.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Immunomodulation of Immune Suppression - Cancers that respond

A

•Cancers that respond very well (40- 50% of the time) to checkpoint blockade include:

-Hodgkin’s disease and melanoma

•Cancers with an intermediate response (15-25% efficacy) include:

-NSCLC, head and neck cancers, bladder and urinary tract cancers, renal cell carcinoma and liver cancer.

•All other types of cancers display a low or negative response to checkpoint blockade therapy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Immunomodulation of Immune Suppression - The Bad News

A
  • Unfortunately, 60-65% of patients exhibit immune related adverse events (AEs) affecting skin, GI tract and endocrine organs. Occasionally, autoimmune disease develops confirming the role of CTLA-4 in the maintenance of self-tolerance. These AEs include enteropathy, autoimmune cytopenias, hemolytic anemia, thyroid disease, arthritis, psoriasis, granulomatous lung disease and lymphocytic infiltration of non-immune organs.
  • Interestingly, and for reasons not completely understood, the commensal microbiome composition of the patient influences the efficacy of anti-PD-1 therapy, as well as anti-CTLA-4 treatment. Cancer itself or the concomitant use of antibiotics may result in microbiome dysbiosis. It appears that certain bacterial strains influence IL-12 production by the innate immune system and migration of T cells into the tumor, both of which will affect therapeutic efficacy.
  • In addition, patient HLA genotype influences success, in that patients that are homogenous at one or more class I loci do significantly worse in terms of outcome.
  • Overall, the fact that checkpoint blockade therapy does indeed enable anti-cancer immune responses in many patients indicates that in many individuals their immune system is capable of recognizing either tumor-specific or self-antigens.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Other Kinds of Antibody-Based Therapy - Mechanisms

A
  1. Agonist action causes direct death of cell
  2. Antagonist prevents signaling, proliferation
  3. Payload delivery of an enzyme or toxin
17
Q

Antibody mediated therapy - Blinatumomab

A

•there is a new class of BiTE antibodies (bispecific T cell engaging) directed against CD19/CD3 that is very effective for treatment of ALL (Blinatumomab by Amgen)

18
Q

Antibody mediated therapy - Rituximab

A
  • Non-Hodgkin’s Lymphoma (a B cell Cancer)
  • Rituximab (Rituxan, IDEC-C2B8)-chimeric (mouse-human) monoclonal antibody directed at CD20 protein on B cells (93% of B cell lymphomas express CD20).
  • Overall response rate (ORR): 48% (6% CR).
  • B cells are killed and cleared by the RES.
19
Q

Antibody mediated therapy - Herceptin

A
  • Breast Cancer
  • Herceptin-monoclonal antibody against Her-2/neu.
  • Herceptin is a recombinant DNA-derived, humanized antibody that binds to extracellular domain of Her-2/neu.
  • The Her-2 protein is overexpressed in 25-30% of primary breast cancers.
  • Herceptin is approved by the Food and Drug Administration for treating metastatic disease in patients who relapsed after prior chemotherapy: (14% ORR)
20
Q

Antibody mediated therapy - Daratumumab

A

•There is a new mAb therapy for multiple myeloma called daratumumab (Darzalex) that targets the CD38 molecule present on plasma cells; FDA approved 2018.

21
Q

Antibody mediated therapy - Alemtuzumab

A
  • CD52
  • CLL
22
Q

Antibody mediated therapy - Bevacizumab

A
  • VEGF
  • Tumor vasculature
23
Q

Antibody mediated therapy - Cetuximab, panitumumab, nimotuzumab, 806

A
  • EGFR
  • glioma
  • lung
  • breast
  • colon
  • head and neck
24
Q

Antibody mediated therapy - Trastuzumab, pertuzumab

A
  • ERBB2 (HER2/neu)
  • breast
  • colon
  • lung
  • ovarian
  • prostate
25
Q

Limitations of Antibody Therapy

A
  • *Heterogeneity of antigen expression on the tumor (either initially, or as therapy proceeds)
  • *Delivery of immunoconjugate low due to low antigen expression on tumors or poor vascularization of tumors
  • *Toxicity: marrow suppression, vascular leak syndrome
  • Therapeutic dose reaching tumor is too small; therapeutic ratio not high enough
  • Development of HAMA (human anti-mouse antigen) responses (humanization via genetic engineering can solve this problem)
  • Lack of absolute specificity of monoclonal antibodies. Too many off-target AEs.
26
Q

Adoptive Cellular Therapy (ACT)

A
  • adoptive cell therapy (ACT) involves the expansion of tumor- specific T (immune) cells in tissue culture in vitro, and then transfer into lympho-depleted patients.
  • The hope is that these T cells will traffic to the tumor and mediate its destruction.
27
Q

Adoptive Cellular Therapy (ACT) - Barriers

A
  • The biggest barrier for T cell immunotherapy is whether the low-avidity T cell population that can recognize self-tumor-associated antigens (having survived self-tolerizing mechanisms), are able to be sufficiently activated to reject established tumors. This limitation may be overcome by genetically engineering T cells to express a given TCR or receptor with high avidity for the tumor antigen.
  • Most ACT therapies are just now being FDA approved, many are in the experimental stage. They are available only at a limited number of clinical trial locations throughout the world, and are still incredibly expensive as they require specialized production facilities and highly trained expertise. Only recently have several (3) ACT approaches received FDA approval (2017).
28
Q

Adoptive Cellular Therapy (ACT) - Lymphokine activated killer (LAK) cells

A

•Autologous peripheral blood mononuclear cells (PBMCs) are cultured with high doses of IL-2, contains a mix of CD4+ and CD8+ T cells, as well as NK cells-the first ACT developed by Rosenberg at NIH-some efficacy vs. melanoma

29
Q

Adoptive Cellular Therapy (ACT) - Tumor infiltrating lymphocytes (TILs)

A
  • Autologous T cells isolated from excised tumors are expanded in culture with IL-2
  • For metastatic melanoma, clinical response rates range from 49-72%, with ~22% of patients showing complete tumor regression, with many still tumor-free up to 57 months post-treatment.
  • TILs from other solid cancers (breast, colon) do not seem as effective – only melanoma currently treated with TILs-also developed by Rosenberg at NIH
30
Q

Adoptive Cellular Therapy (ACT) - Genetic expression of recombinant TCRs

A
  • Autologous T cells are genetically engineered via lentiviruses or retroviruses to express a high affinity TCR specific for tumor antigens (such as via a CRISPR- Cas9 approach)
  • Typically, a high affinity TCR is identified in a patient with a good response to TIL ACT, and then cloned into vectors for expression in T cells from other patients with the same HLA restriction elements (which is a limiting factor).
31
Q

Adoptive Cellular Therapy (ACT) - Chimeric antigen-receptor modified T cells (CAR-T)

A

•Adoptive T cell transfer originally displayed limited efficacy in some patients with malignant melanoma and renal cell carcinoma. Genetic modification of such T cells (usually collected from patient peripheral blood) with chimeric antigen receptors (CARs) directed against common tumor antigens (e.g., CD19, CD20 and CD22 in B cell tumors) has shown greater promise and has recently been FDA-approved for several indications (e.g., pediatric ALL)

32
Q

Adoptive Cellular Therapy (ACT) - Chimeric antigen-receptor modified T cells (CAR-T) are made of…

A
  • The CARs are generally composed on one or more intracellular T cell signaling domains (e.g., CD3-zeta with CD28, 4-1BB, ICOS or OX-40) and an extracellular antigen-binding domain derived from an antibody (scFv).
  • Such a construct generally has a higher affinity than the native TCR and is no longer HLA-restricted in its recognition processes.
33
Q

Adoptive Cellular Therapy (ACT) - Chimeric antigen-receptor modified T cells (CAR-T) Additional Info

A
  • These CAR-T populations persist long term once injected into patients, being measurable for at least a decade.
  • Complete response rates (CRs) have been 70-90% for ALL and 30-50% for patients with CLL and NHL.
  • Response rates appear to last for 5 years or longer in optimal patients.
  • The first approved CAR-T was Kymriah (tisagenlecleucel) from Novartis for ALL which is also now approved for NHL, including adult DLBCL with ORR of 50% and CR of 32%.
  • There has now been another FDA approved CAR-T therapy, termed Yescarta (axicabtagene ciloleucel) from Kite Pharmaceuticals (in 2017).
34
Q

Adoptive Cellular Therapy (ACT) - Chimeric antigen-receptor modified T cells (CAR-T) - Drugs

A
  • Kymriah
  • Yescarta
35
Q

Adoptive Cellular Therapy (ACT) - Chimeric antigen-receptor modified T cells (CAR-T) - The Bad News (CRS)

A
  • Infusion of large numbers of CAR-T cells (109 cells or more) into cancer patients is associated with frequent and significant adverse events, often termed cytokine release syndrome (CRS) and tumor lysis syndrome
  • CRS is a result of secretion of large amounts of interferon-gamma, TNF-alpha, IL-2 and IL-6. Patients often present with fever, hemodynamic compromise, macrophage activation and neurological complications (due to loss of endothelial cell integrity).
  • there is observed long term loss of normal B cell populations which must be managed (often with IVIg therapy) and can be associated with infectious complications due to prolonged hypogammaglobinemia.
36
Q

Treatment for CRS

A
  • Treatment with anti-IL-6 mAbs (tocilizumab) seems to be the most effective therapy for CRS, while treatment with anti-IL6R often results in increased AEs. Severity of the adverse effects seems to correlate with tumor burden and rate of tumor reduction upon infusion.
  • Unfortunately, despite FDA approval for multiple indications, the cost of CAR-T therapy is estimated to be $400,000-$750,000 per patient, with 50% reimbursement.
37
Q

Adoptive Cellular Therapy (ACT) - The Future

A

•NK cells are now also being investigated as a means of adoptive cell therapy with hopes of fewer autoimmune side effects or cytokine release storm symptoms seen with T cells.