Zephyrnet Logo

Emerging immunomodulatory strategies for cell therapeutics

Date:

Modulating the immune response for successful cellular therapies

The convergence of bioengineering innovations with advances in immunology has substantially expanded the landscape of cellular therapies (see Glossary) [

1.

  • Cubillos-Ruiz A.
  • et al.
Engineering living therapeutics with synthetic biology.

]. Cellular therapies aim to treat or manage a disease by introducing living cells that will integrate within the host to restore or eliminate dysfunctional tissues. They typically encompass stem cells (SCs) or non-SCs derived from autologous, allo- or xenogeneic sources, either unaltered or genetically engineered. Currently, hematopoietic SC and chimeric antigen receptor T (CAR-T) cell therapies for hematologic disorders and cancers are the predominant clinically approved products. As of August 2022, there are more than 3000 active clinical trials of cellular therapies. These trials primarily use SCs and blood cells (leukocytes, red blood cells, and platelets) for a spectrum of therapeutic indications such as cancer, hematologic disorders as well as autoimmune, cardiovascular, degenerative, and infectious diseases. A breakdown of the current landscape of active clinical trials of cellular therapies is provided elsewhere [

2.

  • Wang L.L.-W.
  • et al.
Cell therapies in the clinic.

].

Despite this expanding pipeline, host immune response to cellular therapies remains a challenge that could fundamentally impede clinical adoption and desirable outcomes [

3.

  • Petrus-Reurer S.
  • et al.
Immunological considerations and challenges for regenerative cellular therapies.

]. Host immune rejection could occur even for cells of allogeneic origin with human leukocyte antigen (HLA) matching, attributable to mismatched minor alleles [

3.

  • Petrus-Reurer S.
  • et al.
Immunological considerations and challenges for regenerative cellular therapies.

]. Systemic immunosuppression through immunosuppressant drugs is routinely used to reduce rejection. Immunosuppressive treatments are categorized as ‘induction’ (high-intensity immunosuppression immediately post-therapy), ‘maintenance’ (long-term immunosuppression to prevent chronic rejection), and ‘rejection treatment’ (used to treat acute rejection). These immunosuppressants include calcineurin inhibitors (e.g., tacrolimus and cyclosporine), corticosteroids (prednisone), monoclonal antibodies (e.g., basiliximab, adalimumab, and rituximab), inosine monophosphate dehydrogenase inhibitors (mycophenolate mofetil), mechanistic target of rapamycin (mTOR) inhibitor (rapamycin), and depleting antibodies (anti-thymocyte globulins). In general, immunosuppressive agents are administered to target T and B cells, which are key in immune rejection. However, systemic immunosuppression is undesirable due to increased risks of infections, cancers, and organ damage.

Therefore, the clinical success of cellular therapies demands innovations in facilitating as well as maintaining immune acceptance for favorable long-term therapeutic outcome. This review highlights emerging immunomodulatory strategies to attenuate immune rejection or promote tolerance to cellular therapies, with a discussion on local, site-specific immunomodulation measures (Figure 1, Key figure). We exclude solid organ transplantation and tissue grafts, which are extensively reviewed elsewhere [

4.

  • Slepicka P.F.
  • et al.
Harnessing mechanisms of immune tolerance to improve outcomes in solid organ transplantation: a review.

]. We provide a perspective on opportunities for accelerating translation of innovative immunomodulation strategies that synergize with cellular therapies toward achieving widespread clinical success.

Figure 1

Figure 1Key figure. Cellular therapies, administration, and immunomodulatory strategies.

Show full caption

There are various types of cellular therapies (A) and different administration methods and sites (B), and correspondingly, diverse approaches to modulate the immune response (C) to achieve maximal therapeutic benefit. Cellular therapies comprise stem cell (SC) or non-SC-based sources. SC-based therapies include those of embryonic SC (ESC), mesenchymal SC (MSC), or pluripotent SC (PSC) origin. Non-SC-based therapies include blood cells such as dendritic cells (DCs), regulatory T cells (Tregs), and CAR-T cells, and tissue-specific cells. There are different options for administration methods and sites, depending on the type of cellular therapy and therapeutic indication. These sites could have distinct immune composition and necessitate different immunomodulatory strategies for optimal outcome. As immune rejection is a key obstacle for cellular therapy, numerous immunomodulatory interventions have emerged to improve clinical success. Clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR/Cas9) technology is used to disrupt immunogenic cell surface markers such as human leukocyte antigens (HLAs), T cell receptors (TCRs), and co-stimulatory molecules (e.g., CD40) at the genomic level, resulting in immune avoidance or T cell anergy. Similarly, RNA therapeutics, such as RNAi technologies for targeted gene silencing or mRNA for transient protein expression, using nanoparticles (NPs) as delivery vehicles, are leveraged to modulate immune activity. Immunosuppressants can be codelivered with cellular therapy to create a locally immunosuppressed microenvironment. On a similar note, SCs or cells derived from SCs can be used to induce tolerance or avoid host immunoreaction. Abbreviation: CAR-T cells, chimeric antigen receptor T cells.

CRISPR-Cas9 genome editing

Genome editing technologies such as the clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9) system have led to the development of ‘off-the-shelf’, or ‘universal’, engineered cell therapies with little to no immunogenicity. The CRISPR-Cas9 system generates targeted double-strand breaks (DSBs) in the genome. which can be repaired by the cell. For repair, cells can use non-homologous end joining (NHEJ), which can effectively knockout the gene of interest. Alternatively, homology-directed repair (HDR) can occur if a donor DNA template is provided, which allows for targeted insertion of exogenous genes. Depending on the application, gene editing to avoid immune recognition has mainly focused on the elimination of genes encoding for immunogenic surface markers, such as HLAs and T cell receptors (TCRs). For regenerative therapies using iPSCs, the focus has been on deletion of the B2M and CIITA genes required for the expression of HLA class I and II genes, respectively, which typically drive the alloimmune response (Figure 2). Although complete HLA knockout helps to avoid recognition by host CD4+ and CD8+ T cells, HLA-1 deficiency can lead to activation of recipient natural killer (NK) cells and transplant rejection [

5.

  • Duygu B.
  • et al.
HLA class I molecules as immune checkpoints for NK cell alloreactivity and anti-viral immunity in kidney transplantation.

]. Allele-specific editing of polymorphic HLA-1 to express common HLA-C alleles which could be matched to >90% of the world’s population, in addition to HLA-II elimination, was used to generate iPSCs which suppressed recognition by both NK and T cells [

6.

  • Xu H.
  • et al.
Targeted disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility.

]. Alternatively, overexpression of nonpolymorphic HLA-1 molecules such as HLA-E in stem and progenitor cells can also inhibit NK cell lysis activity [

7.

  • Gornalusse G.G.
  • et al.
HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells.

,

8.

  • Sugita S.
  • et al.
Natural killer cell inhibition by HLA-E molecules on induced pluripotent stem cell–derived retinal pigment epithelial cells.

]. Finally, iPSCs have been edited to simultaneously disrupt HLA-1 expression and overexpress CD47, a ‘don’t-eat-me’ signal which effectively inhibits phagocytosis and thus prevents macrophage and NK cell-mediated transplant rejection [

9.

  • Deuse T.
  • et al.
Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients.

]. We further expand on iPSCs in the section ‘Stem cell-derived immunomodulatory therapeutics’.

Figure 2

Figure 2CRISPR- and RNA-based technologies for immunomodulation.

Show full caption

These technologies can be applied to either (A–C) reduce the immunogenicity of cell therapies or (D) induce tolerance toward the cell therapy by the host immune system. Strategies to reduce donor immunogenicity include (A) CRISPR-Cas9 editing of donor cells to express common or nonpolymorphic HLAs or knockout of immunostimulatory HLA gene through CRISPR-Cas9 or RNAi therapies. Additionally, donor cells can also be genetically engineered to (B) overexpress immunosuppressive surface markers such as CD47 or (C) secrete anti-inflammatory cytokines such as IL-10 to modulate the local immune environment. (D) Additionally, recipient tolerance toward allogeneic cell therapies can be achieved through targeted activation of Tregs using mRNA-based IL-2 production or tolerogenic vaccines. Abbreviations: CRISPR-Cas9, clustered regularly interspaced short palindromic repeats-associated protein 9; HLA, human leukocyte antigen; IL, interleukin; NK, natural killer; Tregs, regulatory T cells.

For generating ‘off-the-shelf’ CAR-T cells, human T cells have been edited to eliminate both CD7 and TRAC, with the latter’s deletion blocking TCR-mediated signaling which causes graft-versus-host disease (GVHD) [

10.

  • Cooper M.L.
  • et al.
An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies.

]. These doubly edited CAR-T cells were effective in killing T cell acute lymphoblastic leukemia (T-ALL) in vivo without evidence of xenogeneic GVHD [

10.

  • Cooper M.L.
  • et al.
An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies.

]. Additionally, CAR-T cells edited to lack TCR and HLA-1 reduced alloreactivity and GVHD associated with the cell therapy with a simultaneous third edit to delete PD1 [

11.

  • Ren J.
  • et al.
Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition.

]. The PD1 inhibitory pathway can attenuate CAR-T cell-mediated antitumor activity. As such, the abrogation of the PD1 inhibitory pathway improved antitumor efficacy.

Furthermore, Cas9 was used to endow INS-1E, a rat insulin-secreting β-cell line, with immunomodulatory functions. Specifically, INS-1E was precisely engineered to continuously produce interleukin-10 (IL-10) cytokine in a glucose-responsive manner as the knock-in site was located in the c-peptide region [

12.

  • Lim D.
  • et al.
Engineering designer beta cells with a CRISPR-Cas9 conjugation platform.

]. The continuous local secretion of IL-10 can reduce fibrosis and protect β cells from proinflammatory cytokine-induced cell death, with minimal systemic effect on the host immune system.

The generation and use of genetically engineered cell therapies with minimal immunogenicity come with safety concerns which must be considered. Gene editing with CRISPR-Cas systems often involves DSBs in the genome which can cause unintended large deletions, complex genomic rearrangements, or aneuploidy leading to harmful pathologies if not carefully monitored and addressed [

13.

  • Nahmad A.D.
  • et al.
Frequent aneuploidy in primary human T cells after CRISPR–Cas9 cleavage.

,

14.

  • Kosicki M.
  • et al.
Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements.

]. These risks are especially important to consider in cell therapies engineered to avoid immune recognition because malignant transformation of the engineered cells may escape sensing by host immune cells. Therefore, there is growing interest in using gene editing tools which do not induce DSBs such as base editors and prime editors, or even epigenomic editing tools, to lower immunogenicity of cell therapies [

15.

  • Bashor C.J.
  • et al.
Engineering the next generation of cell-based therapeutics.

].

RNA therapeutics for immune tolerance

RNA therapeutics, such as RNAi technologies, for targeted silencing of genes or in vitro-transcribed mRNA for transient expression of encoded peptides or proteins, hold significant potential in promoting immune tolerance toward cell therapies. Similar to CRISPR-Cas9, RNAi offers the ability to silence the expression of immunogenic alloantigens through mRNA transcript degradation or translation inhibition. This is achieved through the use of short, double-stranded RNA (dsRNAs) in combination with the endogenous effector RNA-induced silencing complex to facilitate homology-directed gene silencing at the post-transcriptional level. Therefore, the elimination of surface MHC molecules can be achieved through RNAi without the risks associated with gene editing as mentioned in the previous section (Figure 2).
For vascularized cell therapies, host immune responses to graft endothelial cells (ECs) expressing nonmatched HLA can lead to transplant rejection. Pretreatment of donor blood vessels ex vivo with siRNA targeting CIITA eliminated HLA-II expression in ECs and prevented rejection of donor arteries by adoptively transferred allogeneic peripheral blood mononuclear cells (PBMCs) in immunodeficient mice [

16.

  • Cui J.
  • et al.
Ex vivo pretreatment of human vessels with siRNA nanoparticles provides protein silencing in endothelial cells.

]. While the use of siRNA is promising for transient knockdown of HLA, which may be beneficial in promoting initial immune tolerance, permanent elimination may be more desirable to improve the likelihood of the long-term viability of cell therapy. Lentiviral delivery of short hairpin RNA (shRNA) targeting B2M can enable stable expression of the interfering RNA to achieve a more permanent knockdown of HLA-1. This approach has been used to generate HLA-1 knocked down cells, which prevents CD8+ T cell response with residual HLA-1 expression preventing NK cell lysis [

17.

  • Figueiredo C.
  • et al.
Class-, gene-, and group-specific HLA silencing by lentiviral shRNA delivery.

]. Stably expressed shRNA targeting B2M has also been used to generate HLA-1 knocked down iPSCs that can be derived into megakaryocytes capable of generating platelets following transfusion into a mouse model for platelet refractoriness [

18.

  • Börger A.-K.
  • et al.
Generation of HLA-universal iPSC-derived megakaryocytes and platelets for survival under refractoriness conditions.

].

With the approval of two mRNA vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and many more mRNA therapies in clinical trials, there is growing interest in the use of mRNA to promote immune tolerance toward cell therapies [

19.

  • Barbier A.J.
  • et al.
The clinical progress of mRNA vaccines and immunotherapies.

]. The focus of mRNA therapies in this area has been on the activation and expansion of regulatory T cells (Tregs) which play an important role in immunosuppression and prevention of GVHD [

20.

  • Guo W.-W.
  • et al.
Regulatory T cells in GVHD therapy.

]. mRNA encoding for human IL-2 mutein was designed to preferentially bind IL-2 receptor α (IL-2Rα) receptors on Tregs and avoid proinflammatory T cell activation [

21.

  • de Picciotto S.
  • et al.
Selective activation and expansion of regulatory T cells using lipid encapsulated mRNA encoding a long-acting IL-2 mutein.

]. Delivery of this mRNA led to Treg activation and expansion in mouse and non-human primate models and effectively reduced acute GVHD in mice. However, the dual role of IL-2 in promoting both Tregs and proinflammatory T cells will require careful monitoring of T cell responses to avoid exacerbating immunogenicity toward cell therapies.

Alternatively, tolerogenic mRNA vaccines may be used to induce alloantigen-specific tolerance. Such vaccines are designed with chemically modified mRNA that are carefully purified to remove dsRNA contaminants. The resulting noninflammatory mRNA vaccines can induce tolerance toward an encoded antigen when presented to T cells in the absence of co-stimulatory molecules [

22.

  • Kariko K.
  • et al.
Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA.

,

23.

  • Kariko K.
  • et al.
Generating the optimal mRNA for therapy: HPLC purification eliminates immune activation and improves translation of nucleoside-modified, protein-encoding mRNA.

,

24.

  • Krienke C.
  • et al.
A noninflammatory mRNA vaccine for treatment of experimental autoimmune encephalomyelitis.

]. Currently, the use of tolerogenic mRNA vaccines has been limited to the induction of autoantigen-specific Treg responses for the prevention of autoimmune disease onset in a mouse model of multiple sclerosis [

24.

  • Krienke C.
  • et al.
A noninflammatory mRNA vaccine for treatment of experimental autoimmune encephalomyelitis.

]. We envision that prophylactic tolerogenic mRNA vaccines encoding for donor HLA could induce tolerance toward HLA-mismatched cell therapies mediated by donor HLA-specific Tregs.

MSCs

MSCs secrete cytokines, chemokines, and growth factors responsible for regulation of inflammation and immune response [

56.

  • Song N.
  • et al.
Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential.

]. MCSs can inhibit allogeneic T cell responses, promote Tregs, trigger DC differentiation into tolDCs, transform proinflammatory M1 macrophages to anti-inflammatory M2 phenotype, as well as inhibit NK cell proliferation. These immunosuppressive properties of MSCs have rendered them attractive for codelivery in cellular therapies, including for that of islets (NCT02384018).

In a murine model of retinal degenerative disease, cotransplantation of MSCs with fetal retinal pigment epithelial (RPE) cells suppressed host immunoreaction, allowing prolonged graft survival for preserving retina function [

57.

  • Pan T.
  • et al.
Combined transplantation with human mesenchymal stem cells improves retinal rescue effect of human fetal RPE cells in retinal degeneration mouse model.

]. The co-encapsulation of hepatocyte nuclear factor-4 alpha (HNF4α) overexpressing MSCs with hepatocytes promoted M2 macrophage polarization and alleviated inflammation in an acute liver failure murine model [

58.

  • Kong D.
  • et al.
Co-encapsulation of HNF4α overexpressing UMSCs and human primary hepatocytes ameliorates mouse acute liver failure.

]. Yoshida et al. showed that syngeneic MSCs induced immune tolerance to iPSC-derived cardiomyocytes by promoting Tregs and triggering CD8+ T cell apoptosis [

59.

  • Yoshida S.
  • et al.
Syngeneic mesenchymal stem cells reduce immune rejection after induced pluripotent stem cell-derived allogeneic cardiomyocyte transplantation.

]. The combination of iPSC-derived cardiomyocytes and MSCs yielded improved cardiac function in a murine model of myocardial infarction, compared with single-cell population transplant alone [

60.

  • Neef K.
  • et al.
Co-transplantation of mesenchymal stromal cells and induced pluripotent stem cell-derived cardiomyocytes improves cardiac function after myocardial damage.

]. In the context of diabetes, good manufacturing practice-compliant human umbilical cord perivascular MSCs cotransplantation with islets in diabetic mice achieved T cell suppression and maintenance of tight glycemic control [

61.

  • Forbes S.
  • et al.
Human umbilical cord perivascular cells improve human pancreatic islet transplant function by increasing vascularization.

]. Furthermore, MSCs engineered to express PD-L1/CTLA4-Ig (eMSCs) can induce local immunosuppression and support allogeneic rat islet engraftment without systemic immunosuppression [

62.

  • Wang X.
  • et al.
Engineered immunomodulatory accessory cells improve experimental allogeneic islet transplantation without immunosuppression.

].

Despite their promise, challenges with SCs include challenges with achieving full functional maturation, unclear long-term fate, immunogenicity, and cost and complexity of large-scale manufacturing. On that note, quality control between different SC sources, ease of procurement, and upscaling while maintaining stable phenotype are important criteria for clinical translatability [

63.

  • Levy O.
  • et al.
Shattering barriers toward clinically meaningful MSC therapies.

]. We refer the readers to the section ‘Concluding remarks and future perspectives, for considerations regarding clinical translation.

Concluding remarks and future perspectives

The wide breadth of investigations in the field are exemplified by the numerous clinical trials exploring immunomodulatory strategies for cell therapy, some of which are listed in Table 1. The first-in-human and open-label, clinical trial of CAR-Tregs for inducing and maintaining immune tolerance to kidney transplants was initiated in 2021 (NCT04817774). Recently, a clinical trial in Japan suggested that cord blood transplantation combined with intra-BM injection of MSCs could prevent GVHD without engraftment inhibition [

75.

  • Goto T.
  • et al.
Phase I clinical trial of intra-bone marrow cotransplantation of mesenchymal stem cells in cord blood transplantation.

]. Further, some innovative immunomodulatory interventions include clinical investigations of CRISPR-Cas9- and mRNA-based therapeutics to mitigate immune rejection or promote tolerance (NCT05210530).

Table 1Selected clinical trials involving cell therapy and immunomodulation for regenerative therapy

a

Abbreviations: ADSCs, adipose-derived stem cells; BM-MSCs, bone marrow mesenchymal stem cells; COPD, chronic obstructive pulmonary disease; hESC-RPE, human embryonic stem cell-derived retinal pigmented epithelium; Ph1, Phase 1; Ph2, Phase 2; SPIONs, superparamagnetic iron oxide; thyTreg, thymus-derived Tregs.

As with all novel biomedical technologies, safety and ethical concerns must be addressed prior to clinical translation. Further, critical considerations for translation of cellular therapies include reproducibility, large-scale production [

76.

Stem-cell start-ups seek to crack the mass-production problem.

], and standardization and quality control protocols [

77.

  • Fritsche E.
  • et al.
Toward an optimized process for clinical manufacturing of CAR-Treg cell therapy.

,

78.

  • Pratt C.B.
  • Alexander M.
Importance of CAR-T cell therapy monitoring using high-throughput assays.

] (see Outstanding questions). To resolve these challenges, various public and private programs have established fundamental guidelines for good manufacturing processes in the biofabrication industry. A relevant example of these efforts is the BioFabUSA program established at the Advanced Regenerative Manufacturing Institute (ARMI). BioFabUSA is a public–private partnership consisting of industry, academia, government, and nonprofit organizations. This unique partnership focuses on directing science and engineering resources toward enabling scalable, consistent, and cost-effective manufacturing of cellular therapies. Within this, advances in robotics, information technology, computational sciences, and artificial intelligence infrastructures [

79.

  • Tucker B.A.
  • et al.
Autologous cell replacement: a noninvasive AI approach to clinical release testing.

] will be fundamental to rendering cellular therapies more personalized, accessible, and affordable. In addition, as cellular therapies are essentially living drugs, mastering the supply chain from appropriate temperature regulated transportation logistics and storage to proper thawing and administration is important for widescale implementation in a reproducible manner.

Another significant challenge is developing safe and effective delivery strategies for cell therapeutics. Transplantation of pancreatic islets or SC-derived β cells provides a clear example of the importance of the delivery approach on the viability and function of the graft. Despite more than 70 years of research and development in the field, the ideal technological solution for the delivery of these cells has yet to be identified. To this end, novel discoveries in biomaterials and nanomedicine will continue to support these efforts in providing new molecular and cell engineering tools [

80.

  • Cifuentes-Rius A.
  • et al.
Inducing immune tolerance with dendritic cell-targeting nanomedicines.

,

81.

  • Madl C.M.
  • et al.
Bioengineering strategies to accelerate stem cell therapeutics.

].

Finally, new research opportunities reside in developing effective strategies to track cell therapeutics upon delivery in the body [

82.

  • Bulte J.W.M.
  • Daldrup-Link H.E.
Clinical tracking of cell transfer and cell transplantation: trials and tribulations.

], noninvasively monitor their viability and function, as well as modulate immunological response ad hoc. Here, innovative real-time imaging technologies and optogenetic approaches to manipulate cells using light stimuli at specific wavelengths may pave the way for novel discoveries [

83.

  • Bansal A.
  • et al.
Towards translational optogenetics.

].

These translational challenges are massive and require the convergence of multidisciplinary expertise and capabilities. As exemplified by the global response to the SARS-CoV-2 pandemic via the collective actions of academia and industry, which resulted in the ultra-rapid vaccine development and regulatory approval, concerted efforts could enable cellular therapies to reach their full potential.

Outstanding questions

Will the added factor of immunomodulation for cellular therapies, which already require laborious, large-scale manufacturing with rigorous quality standards, impact commercialization costs and be economically feasible? Will genetic cell manipulation alone allow for complete graft immune evasion? What types of gene manipulation are most feasible for a wide spectrum of cell therapeutics? Will potential genotoxicity caused by traditional CRISPR-Cas9 gene editing push the field toward newer CRISPR-based technologies or alternative gene editing approaches? Will tolerogenic vaccines improve transplant outcomes and what will be the key alloantigen(s) to target for vaccination? Will nanotheranostics and optogenetics better support investigation and monitoring of localized immunomodulation? How to better identify if acute rejection is due to the cell therapy or other factors?

Glossary

Allogeneic

cells or tissues derived from a donor of the same species.

Alloimmune response

an immune response to non-self antigens (‘alloantigens’) from members of the same species. An alloimmune response can result in graft rejection.

Apoptosis

a type of programmed cell death leading to self-destruction of cells, triggered by the presence or absence of certain stimuli.

Autoimmunity

immune response against an individual’s own cells or tissues through the presence of antibodies and T lymphocytes directed against self-antigens.

Autologous

cells or tissues derived from the same individual into whom they are transplanted.

Cellular therapy

living cells used for therapy.

Chimeric antigen receptor T (CAR-T) cell

a form of immunotherapy using T cells genetically modified to have a synthetic receptor that binds to a specific target (cancer cells) and mediate immune destruction. They are referred to as ‘chimeric’ because both antigen-binding and T cell activating functions are combined within a single receptor.

Clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9)

a genome editing technology adapted from bacteria that can be used to specifically edit DNA at precise locations. A specially designed RNA molecule is used to guide Cas9 enzyme to a targeted sequence of DNA. Cas9 cuts the targeted DNA sequence for removal, thus allowing for deletion or the addition of a new, customized DNA sequence. CRISPR-Cas9 technology holds promise for treating and preventing previously untreatable diseases such as neurodegenerative, genetic, or hereditary disorders, HIV, and cancer.

Double-strand break (DSB)

occurs when both strands of DNA are cleaved by damaging agents such as ionizing radiation or certain chemicals.

Graft-versus-host disease (GVHD)

GVHD is a life-threatening systemic inflammatory complication that can occur after transplantation when donor T cells in the transplant attack the recipient.

Homology-directed repair (HDR)

a mechanism used by the cell to repair DSBs in DNA, relying on a homologous sequence of DNA, primarily occurring during G2 and S phases of the cell cycle.

Human leukocyte antigen (HLA)

HLA genes code for cell surface proteins in MHCs, which are unique to the individual. HLAs are used by the immune system to differentiate between self and non-self.

Immune privilege

refers to certain sites in the body that are isolated from the immune system, which can tolerate foreign antigens, cells, or tissues without inducing an inflammatory immune response that can lead to rejection.

Immune tolerance

unresponsiveness of the immune system to a specific antigen or a previously encountered antigen. Transplant tolerance refers to the lack of immune responses to antigens from donor cells or grafts, which prevents immune rejection, while reactivity to other antigens remains intact.

Immunosuppressants

agents used to suppress the immune system to prevent the cells from attacking donor cells, which are seen as foreign to the host.

Immunosuppression

suppression of the body’s immune system and consequently, the ability to fight infection and disease. Immunosuppression can be induced by drugs or specialized cells, as well as occur as a result of a disease state.

iPSCs

cells that are obtained by reprogramming terminally differentiated adult cells, such as skin cells, into an embryonic-like pluripotent state to be used as an unlimited source for therapeutic purposes. iPSCs can be created from cells of the same individual who will receive the transplant.

Micelles

spherical amphiphilic structures containing a hydrophobic core and a hydrophilic shell. They are used as drug carriers, as the hydrophilic shell renders micelles water soluble, whereas the hydrophobic core carries the payload.

Microgel

3D network of polymer microfilaments comprising natural or synthetic materials that can be crosslinked using physical, chemical, or light-mediated mechanisms. Microgels are hydrogels with particle sizes bigger than 100 nm and smaller than 100 μm. Microgels are used in biomedical applications such as drug delivery, regenerative medicine, and tissue engineering. For example, microgels can be used to encapsulate drugs and engineered to swell or degrade in response to stimuli such as temperature, pH, and light, for drug release.

Natural killer (NK) cells

cytotoxic lymphocytes of the innate immune system, which are early cellular responders to infected cells or cancers. These cells play a central role in modulating alloimmune responses.

Non-homologous end joining (NHEJ)

the primary pathway to repair DNA DSBs, involving ligation of broken strands, throughout the cell cycle. NHEJ has a higher capacity for repair and is faster than HDR and does not need a repair template.

RNAi

RNAi is a process that triggers sequence-specific suppression of gene expression using double-stranded RNA, either via translational or transcriptional repression.

Stem cells (SCs)

cells that have the ability to self-renew and differentiate into different specialized cell types. SCs can propagate indefinitely and thus be an unlimited source for replacing lost or diseased tissues.

Xenogeneic

cells or tissues derived from a donor from a different species.

spot_img

Latest Intelligence

spot_img