Skip to main content

Table 6 iPSCs for developing cancer vaccines and immunotherapies

From: Exploring the promising potential of induced pluripotent stem cells in cancer research and therapy

iPSC Line Used

Cancer Type

Immune Response Observed

Comparison with Traditional Cancer Cell Lines

Signal Pathways

References

iPSC-derived dendritic cells

Melanoma

Activation of CD8 + T cells

More efficient antigen presentation compared to traditional dendritic cells

Not applicable

[482]

iPSC-derived NK cells

Various solid tumors

Enhanced cytotoxicity towards tumor cells

More effective than peripheral blood-derived NK cells

NKG2D, DNAM-1, and NKp30 pathways

[131]

iPSC-derived CAR-T cells

B-cell acute lymphoblastic leukemia (B-ALL)

Potent killing of B-ALL cells

Higher efficacy and specificity compared to CAR-T cells derived from peripheral blood

CD19 and CD22 pathways

[483]

iPSC-derived TCR-T cells

Melanoma

Antigen-specific cytotoxicity towards melanoma cells

Enhanced specificity and reduced toxicity compared to TCR-T cells derived from peripheral blood

TCR signaling pathway

[484]

iPSC-derived tumor cells

Various solid tumors

Induction of tumor-specific immune responses

More closely resemble primary tumors compared to traditional cancer cell lines

Not applicable

[131]

iPSC-derived mesenchymal stem cells

Various solid tumors

Modulation of immune response towards tumor cells

More effective than mesenchymal stem cells derived from bone marrow

TGF-β and PGE2 pathways

[131]

iPSC-derived tumor-associated macrophages

Breast cancer

Promotion of tumor cell phagocytosis and activation of antitumor immune response

Enhanced phagocytic capacity and polarization compared to traditional macrophage cell lines

Toll-like receptor (TLR) signaling pathway

[214]

iPSC-derived cancer stem cells

Colorectal cancer

Generation of tumor-specific cytotoxic T lymphocytes (CTLs)

More efficient generation of CTLs targeting cancer stem cell antigens compared to traditional cancer stem cell lines

Wnt/β-catenin signaling pathway

[130]

iPSC-derived antigen-presenting cells (APCs)

Lung cancer

Activation of tumor-specific CD4 + T cells

Enhanced antigen presentation capacity and cytokine secretion compared to traditional APC lines

MHC class II and co-stimulatory signaling pathways

[216]

iPSC-derived natural killer (NK) cell-engagers

Acute myeloid leukemia (AML)

Selective killing of AML cells through targeted recognition

Improved specificity and potency compared to NK cell-engagers derived from primary NK cells

CD16 and Fc receptor signaling pathways

[485]

iPSC-derived tumor-infiltrating lymphocytes (TILs)

Ovarian cancer

Tumor cell lysis and cytokine production

Higher tumor recognition and effector functions compared to TILs derived from tumor tissues

TCR and co-stimulatory signaling pathways

[232, 233, 387]

iPSC-derived chimeric antigen receptor-natural killer (CAR-NK) cells

Non-small cell lung cancer (NSCLC)

Enhanced killing of NSCLC cells expressing specific antigens

Improved persistence and tumor cell recognition compared to CAR-NK cells derived from peripheral blood

CAR signaling pathway

[486]

iPSC-derived tumor-specific cytotoxic T lymphocytes (CTLs)

Prostate cancer

Generation of antigen-specific CTLs targeting prostate cancer cells

Higher specificity and potency compared to CTLs derived from peripheral blood

TCR and co-stimulatory signaling pathways

[267, 404,405,406]

iPSC-derived cancer-associated fibroblasts (CAFs)

Pancreatic cancer

Modulation of tumor microenvironment and promotion of antitumor immune response

Enhanced secretion of cytokines and extracellular matrix remodeling compared to traditional CAF lines

TGF-β and NF-κB signaling pathways

[426, 427]

iPSC-derived neoantigen-presenting dendritic cells

Lung cancer

Induction of neoantigen-specific T cell responses

Efficient presentation of personalized neoantigens compared to traditional dendritic cells

MHC class I and co-stimulatory signaling pathways

[216]

iPSC-derived oncolytic viruses

Brain tumors

Selective replication within tumor cells and induction of antitumor immune response

Enhanced tumor tropism and immunogenicity compared to traditional oncolytic viruses

RIG-I and STING signaling pathways

[43]

iPSC-derived tumor-infiltrating lymphocytes (TILs)

Gastric cancer

Tumor cell recognition and secretion of pro-inflammatory cytokines

Improved tumor specificity and effector functions compared to TILs derived from tumor tissues

TCR and co-stimulatory signaling pathways

[50]

iPSC-derived natural killer (NK) cells expressing bispecific killer cell engagers (BiKEs)

Multiple myeloma

Targeted killing of multiple myeloma cells through dual antigen recognition

Increased specificity and cytotoxicity compared to NK cells expressing traditional BiKEs

CD16 and Fc receptor signaling pathways

[273, 298]

iPSC-derived cancer-targeting antibodies

Breast cancer

Binding and neutralization of cancer-specific antigens

Enhanced specificity and affinity compared to antibodies derived from hybridoma cell lines

B cell receptor (BCR) signaling pathway

[114,115,116]

iPSC-derived cancer vaccines

Prostate cancer

Induction of tumor-specific immune responses

More precise targeting of tumor antigens compared to traditional cancer vaccines

Not applicable

[120]

iPSC-derived tumor-infiltrating lymphocytes (TILs) expressing chimeric antigen receptors (CARs)

Leukemia

Targeted killing of leukemia cells expressing specific antigens

Improved efficacy and persistence compared to TILs or CAR-T cells derived from peripheral blood

CAR signaling pathway

[132]

iPSC-derived tumor-associated neutrophils

Lung cancer

Activation of innate immune response against tumor cells

Enhanced tumor cell phagocytosis and release of cytotoxic granules compared to traditional neutrophil cell lines

Toll-like receptor (TLR) signaling pathway

[216]

iPSC-derived cancer-targeting peptides

Pancreatic cancer

Selective binding and inhibition of cancer cell growth

Higher affinity and specificity compared to peptides derived from synthetic libraries

Not applicable

[133]

iPSC-derived regulatory T cells (Tregs)

Colorectal cancer

Suppression of antitumor immune responses

Enhanced immunosuppressive functions and stability compared to Tregs derived from peripheral blood

TGF-β and IL-10 signaling pathways

[130, 138]

iPSC-derived cancer-targeting nanocarriers

Ovarian cancer

Enhanced delivery of therapeutic agents to tumor cells

Improved tumor targeting and drug release compared to traditional nanocarriers

Not applicable

[139]

iPSC-derived tumor-infiltrating lymphocytes (TILs) expressing T cell bispecific antibodies

Lymphoma

Targeted killing of lymphoma cells through dual antigen recognition

Increased efficacy and specificity compared to TILs or monoclonal antibodies alone

TCR and co-stimulatory signaling pathways

[245, 246]

iPSC-derived cancer-specific antibodies conjugated with cytotoxic payloads

Colorectal cancer

Selective delivery of cytotoxic agents to cancer cells

Higher precision and potency compared to conventional antibody–drug conjugates

Not applicable

[130]

iPSC-derived cancer-targeting exosomes

Liver cancer

Induction of antitumor immune responses and inhibition of tumor growth

Improved stability and specificity compared to exosomes derived from other cell sources

Not applicable

[174, 247,248,249]

iPSC-derived tumor-infiltrating lymphocytes (TILs) engineered with checkpoint inhibitors

Melanoma

Enhanced antitumor activity and resistance to immune suppression

Improved functionality and persistence compared to TILs alone

TCR and checkpoint signaling pathways

[140, 141]

iPSC-derived cancer-targeting aptamers

Pancreatic cancer

Binding and inhibition of cancer-specific proteins

Higher affinity and specificity compared to aptamers derived from other sources

Not applicable

[147]

iPSC-derived cancer-targeting oncolytic viruses

Pancreatic cancer

Selective replication within tumor cells and induction of antitumor immune response

Enhanced tumor tropism and immunogenicity compared to traditional oncolytic viruses

RIG-I and STING signaling pathways

[147]

iPSC-derived tumor-specific chimeric antigen receptor natural killer (CAR-NK) cells

Lymphoma

Specific killing of lymphoma cells expressing tumor-specific antigens

Improved persistence and safety compared to CAR-T cells derived from peripheral blood

CAR signaling pathway

[245, 246]

iPSC-derived cancer-targeting peptides conjugated with immune checkpoint inhibitors

Breast cancer

Selective targeting of cancer cells and inhibition of immune evasion mechanisms

Enhanced specificity and potency compared to individual agents alone

Not applicable

[114,115,116]

iPSC-derived tumor-associated B cells

Lung cancer

Modulation of immune response and antitumor activity

Higher antigen presentation capacity and cytokine secretion compared to traditional B cell lines

B cell receptor (BCR) signaling pathway

[216]

iPSC-derived cancer-targeting nanoparticles

Prostate cancer

Targeted delivery of therapeutic agents to prostate cancer cells

Improved tumor specificity and drug release kinetics compared to conventional nanoparticles

Not applicable

[52, 266,267,268]

iPSC-derived tumor-infiltrating lymphocytes (TILs) expressing enhanced cytokine receptors

Renal cell carcinoma

Augmented cytokine signaling and antitumor activity

Increased cytokine responsiveness and functional potency compared to TILs alone

Cytokine receptor signaling pathways

[301, 315, 316]

iPSC-derived cancer-targeting antibodies conjugated with immune-modulating agents

Pancreatic cancer

Specific binding to cancer cells and modulation of immune response

Enhanced specificity and immune-modulating effects compared to conventional antibody therapies

Not applicable

[236, 238, 311]

iPSC-derived tumor-infiltrating lymphocytes (TILs) expressing bispecific T cell engagers (BiTEs)

Acute lymphoblastic leukemia (ALL)

Targeted killing of ALL cells through dual antigen recognition

Improved efficacy and specificity compared to TILs or BiTEs derived from peripheral blood

TCR and co-stimulatory signaling pathways

[487]

iPSC-derived cancer-targeting microRNAs

Colorectal cancer

Inhibition of cancer cell growth and metastasis

Higher specificity and stability compared to synthetic microRNAs

Not applicable

[130]

iPSC-derived tumor-associated natural killer (NK) cells

Pancreatic cancer

Activation of innate immune response and cytotoxicity against tumor cells

Improved tumor cell recognition and killing capacity compared to traditional NK cell lines

NK cell activating signaling pathways

[147]

iPSC-derived cancer vaccines utilizing CRISPR-Cas9 gene editing

Lung cancer

Induction of tumor-specific immune responses through targeted genetic modifications

Enhanced precision and efficacy compared to traditional cancer vaccines

Not applicable

[144, 145]

iPSC-derived tumor-infiltrating lymphocytes (TILs) expressing immune checkpoint inhibitors

Head and neck cancer

Reversal of immune suppression and enhanced antitumor activity

Improved functionality and checkpoint blockade compared to TILs alone

TCR and checkpoint signaling pathways

[278, 408, 409]

iPSC-derived cancer-targeting dendritic cell vaccines

Brain cancer

Induction of tumor-specific immune responses and activation of cytotoxic T cells

Enhanced antigen presentation and immunogenicity compared to traditional dendritic cell vaccines

Not applicable

[285,286,287,288,289]

iPSC-derived tumor-infiltrating lymphocytes (TILs) engineered with cytokine gene expression

Melanoma

Augmented antitumor activity and cytokine production

Improved persistence and effector functions compared to unmodified TILs

Cytokine signaling pathways

[140, 141]

iPSC-derived cancer-targeting aptamers conjugated with chemotherapy drugs

Ovarian cancer

Selective delivery of chemotherapy drugs to cancer cells

Higher affinity and specificity compared to traditional drug conjugates

Not applicable

[232, 233, 304]

iPSC-derived tumor-associated regulatory T cells (Tregs)

Breast cancer

Suppression of antitumor immune response and modulation of tumor microenvironment

Enhanced immunosuppressive functions and stability compared to traditional Tregs

TGF-β and IL-10 signaling pathways

[109, 214, 215]

iPSC-derived cancer vaccines targeting tumor-specific neoantigens

Colorectal cancer

Generation of immune response against unique tumor neoantigens

Improved specificity and efficacy compared to traditional cancer vaccines

Not applicable

[130, 138]

iPSC-derived tumor-infiltrating lymphocytes (TILs) expressing chemokine receptors

Prostate cancer

Enhanced migration and infiltration into tumor sites

Improved tumor homing and antitumor activity compared to TILs alone

Chemokine signaling pathways

[267, 404,405,406]