Advancements in Gene-Edited Islet Cell Transplants for Type 1 Diabetes: A Comprehensive Review

An Advanced Review of Gene-Edited Islet Cell Transplants for Type 1 Diabetes: Mechanisms, Clinical Progress, and Future Prospects

Many thanks to our sponsor Esdebe who helped us prepare this research report.

Abstract

Type 1 Diabetes (T1D) stands as a chronic autoimmune disorder marked by the specific destruction of insulin-producing pancreatic beta-cells, culminating in profound insulin deficiency and persistent hyperglycemia. The longstanding therapeutic cornerstone, exogenous insulin therapy, although life-sustaining, fails to replicate physiological glucose homeostasis and presents significant challenges, including the risk of acute hypoglycemic events and the long-term burden of microvascular and macrovascular complications. Pancreatic islet cell transplantation emerged as a promising alternative, offering the potential to restore endogenous insulin production and achieve normoglycemia. However, its widespread applicability has been severely constrained by two principal barriers: the critical scarcity of suitable donor organs and the unavoidable necessity for lifelong systemic immunosuppression to prevent allograft rejection, which itself carries substantial risks of opportunistic infections, malignancies, and renal dysfunction.

The advent of revolutionary gene-editing technologies, most notably the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 system, has catalyzed a paradigm shift in transplantation biology. This powerful molecular tool enables precise and efficient genomic modifications, opening unprecedented avenues for engineering donor islet cells to achieve robust immune evasion. Such ‘hypoimmune’ islet cells hold the transformative potential to negate the requirement for chronic immunosuppression and, particularly when derived from stem cells, to overcome the persistent shortage of donor material.

This comprehensive review meticulously examines the current landscape of gene-edited islet cell transplants for T1D. It delves deeply into the intricate molecular and cellular mechanisms underpinning immune evasion strategies, critically evaluates the outcomes of pioneering clinical trials, scrutinizes the multifaceted challenges related to scalability and manufacturing, navigates the complex ethical and societal considerations inherent in genomic manipulation, and provides a detailed comparative analysis with traditional transplantation methodologies. By synthesising cutting-edge research and clinical developments, this report aims to illuminate the profound promise and ongoing complexities of gene-edited islet cell therapy as a potential curative strategy for T1D.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

1. Introduction

Type 1 Diabetes Mellitus (T1DM), affecting millions globally, is an autoimmune disease driven by the selective destruction of the insulin-secreting beta-cells within the pancreatic islets of Langerhans. This relentless attack leads to an absolute deficiency of insulin, a hormone critical for glucose metabolism, resulting in hyperglycemia and a cascade of severe complications if left inadequately managed. These complications encompass debilitating microvascular damage (retinopathy, nephropathy, neuropathy) and life-threatening macrovascular events (cardiovascular disease, stroke), which significantly impair patient quality of life and shorten life expectancy [11].

For decades, the primary therapeutic approach has been the exogenous administration of insulin, delivered via multiple daily injections or continuous subcutaneous infusion pumps. While insulin therapy has dramatically transformed the prognosis for T1D patients, it remains a symptomatic treatment rather than a cure. Achieving consistently optimal glycemic control, defined as maintaining blood glucose levels within a narrow physiological range, is notoriously challenging. Patients frequently grapple with the threat of hypoglycemia, a potentially fatal complication, and the persistent burden of daily self-management, which impacts their psychological well-being and overall quality of life [12]. Despite rigorous insulin regimens, many individuals still experience long-term complications, underscoring the pressing need for therapies that restore endogenous, regulated insulin production.

Pancreatic transplantation and, subsequently, isolated pancreatic islet cell transplantation, emerged as potential curative strategies. Whole pancreas transplantation, while capable of establishing insulin independence, is a major surgical procedure associated with significant morbidity and mortality, typically reserved for T1D patients with end-stage renal disease also receiving a kidney transplant [13]. Islet cell transplantation, pioneered and refined by protocols such as the landmark Edmonton Protocol, represented a less invasive alternative, demonstrating the capacity to restore physiological insulin secretion and reduce or eliminate the need for exogenous insulin [14]. However, both approaches are fundamentally limited by two critical factors: the severe scarcity of high-quality donor pancreata, which restricts accessibility to a small fraction of eligible patients, and the absolute requirement for lifelong systemic immunosuppressive drug regimens. These powerful medications, essential to prevent immune-mediated graft rejection, carry a heavy toll of adverse effects, including increased susceptibility to infections, nephrotoxicity, cardiovascular complications, and an elevated risk of certain malignancies, often negating the metabolic benefits of transplantation over the long term [15].

The landscape of therapeutic innovation for T1D has been dramatically reshaped by the advent of advanced gene-editing technologies. Among these, the CRISPR/Cas9 system has rapidly risen to prominence due to its unparalleled precision, efficiency, and versatility in making targeted modifications to the genome. This revolutionary tool offers a transformative opportunity to overcome the historical limitations of islet transplantation. By precisely altering the genetic makeup of donor islet cells – or, increasingly, stem cell-derived beta-cells – it is theoretically possible to render them ‘invisible’ or ‘tolerated’ by the recipient’s immune system, thereby eliminating the need for chronic immunosuppression. Furthermore, when combined with the scalability offered by pluripotent stem cell technologies, gene editing presents a viable pathway to generate an inexhaustible supply of universal, immune-evasive beta-cells, addressing both donor scarcity and immune rejection simultaneously. This report seeks to provide a detailed, current overview of this rapidly evolving and highly promising field.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

2. CRISPR/Cas9 Gene-Editing Technology: A Foundational Enabler

CRISPR/Cas9 has revolutionized molecular biology and holds immense promise for therapeutic applications, including the engineering of cells for transplantation. Its power lies in its ability to precisely target and modify specific DNA sequences within a genome [16].

2.1. Origins and Basic Mechanism

The CRISPR/Cas system was originally identified as a bacterial adaptive immune system, providing prokaryotes with a defense mechanism against invading viruses and plasmids. When a bacterium is infected by a phage, it captures snippets of the viral DNA and integrates them into its own genome at specific CRISPR loci. These integrated sequences, known as ‘spacers’, are transcribed into short CRISPR RNAs (crRNAs) [17]. These crRNAs then associate with tracrRNAs (trans-activating CRISPR RNA) and Cas (CRISPR-associated) proteins, forming a ribonucleoprotein complex. The crRNA acts as a guide, leading the Cas protein to complementary sequences in subsequent invading viral DNA, where the Cas protein cleaves and inactivates the viral genome.

The most commonly utilized and best-characterized CRISPR/Cas system for gene editing in eukaryotes is the Type II system from Streptococcus pyogenes, involving the Cas9 nuclease. In this engineered system, the crRNA and tracrRNA are fused into a single synthetic guide RNA (sgRNA). The sgRNA, typically 20 nucleotides long, is complementary to the target DNA sequence, and a Cas9 nuclease acts as molecular scissors. The mechanism proceeds as follows:

  1. Target Recognition: The sgRNA guides the Cas9 enzyme to a specific genomic locus via Watson-Crick base pairing between the sgRNA and the target DNA strand. This binding requires a short protospacer adjacent motif (PAM sequence, typically NGG for SpCas9) immediately downstream of the target sequence in the DNA, which is essential for Cas9 binding and cleavage specificity [18].
  2. DNA Cleavage: Once bound and the target sequence verified by the PAM, the Cas9 nuclease induces a double-strand break (DSB) approximately 3-4 base pairs upstream of the PAM sequence. This DSB is the fundamental event that initiates genome editing.
  3. DNA Repair and Editing Outcome: Eukaryotic cells possess two primary endogenous pathways to repair DSBs:
    • Non-Homologous End Joining (NHEJ): This is the predominant repair pathway and is error-prone. It ligates the broken DNA ends directly, often introducing small insertions or deletions (indels) at the repair site. If these indels occur within a gene’s coding sequence, they can lead to frameshift mutations, premature stop codons, and ultimately result in a functional gene knockout, which is a common strategy for deleting specific genes like HLA [19].
    • Homology-Directed Repair (HDR): This pathway is less frequent and occurs primarily during the S and G2 phases of the cell cycle. HDR requires a homologous DNA template to guide repair. If an exogenous DNA template (donor template) containing desired modifications (e.g., an inserted gene, a precise nucleotide change) is provided along with the CRISPR/Cas9 components, HDR can be harnessed to precisely introduce these desired genetic alterations, such as inserting immunomodulatory genes or correcting specific mutations [20].

2.2. Advantages over Previous Gene-Editing Technologies

Before CRISPR/Cas9, earlier gene-editing tools like Zinc Finger Nucleases (ZFNs) and Transcription Activator-Like Effector Nucleases (TALENs) were developed. While effective, they were significantly more complex and costly to design and implement. ZFNs and TALENs require de novo protein engineering for each new target site, a laborious and time-consuming process. In contrast, CRISPR/Cas9’s specificity is dictated by a short RNA molecule (the sgRNA), which can be easily synthesized and modified. This RNA-guided mechanism makes CRISPR/Cas9 highly adaptable, cost-effective, and amenable to high-throughput and multiplexed editing (simultaneously targeting multiple genes), capabilities crucial for complex cellular engineering required for immune evasion strategies [21].

2.3. Delivery Methods for CRISPR Components

The efficacy of gene editing in therapeutic contexts relies heavily on efficient and safe delivery of the Cas9 nuclease and sgRNA to the target cells. Delivery methods can be broadly categorized into viral and non-viral:

  • Viral Vectors: These leverage the natural ability of viruses to infect cells and deliver genetic material. Commonly used viral vectors include Adeno-Associated Viruses (AAVs) and Lentiviruses. AAVs are generally preferred for in vivo applications due to their low immunogenicity and stable gene expression, while lentiviruses are efficient at transducing dividing and non-dividing cells, making them suitable for ex vivo cell engineering. However, concerns about immunogenicity, insertional mutagenesis (for integrating viruses like lentiviruses), and viral packaging capacity limit their application [22].
  • Non-Viral Methods: These include electroporation, lipid nanoparticles (LNPs), and cell-penetrating peptides. For ex vivo editing of cells like pancreatic islets or stem cell-derived beta-cells, electroporation of Cas9 mRNA or Cas9 ribonucleoproteins (RNP, pre-assembled Cas9 protein and sgRNA) has proven highly effective. RNP delivery offers advantages such as transient expression of Cas9 (reducing off-target effects and immunogenicity from prolonged Cas9 presence), faster editing, and no risk of viral integration [23]. Lipid nanoparticles, currently successful in mRNA vaccine delivery, are also being explored for in vivo delivery of CRISPR components.

In the context of islet cell engineering, the ex vivo approach, where cells are edited in the laboratory and then transplanted, is predominantly favored. This allows for stringent quality control, verification of successful editing, and removal of unedited or off-target edited cells before transplantation, enhancing safety and efficacy.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

3. Mechanisms of Immune Evasion in Gene-Edited Islet Cells

Successfully achieving immune evasion in transplanted islet cells requires a multifaceted approach to circumvent both innate and adaptive arms of the recipient’s immune system. The ultimate goal is to render the transplanted cells ‘hypoimmune’ – capable of functioning without eliciting a destructive immune response. This involves strategic gene modifications to mitigate key immune recognition pathways.

3.1. Understanding Immune Rejection in Transplantation

To appreciate the strategies for immune evasion, it is crucial to understand the intricate mechanisms of allograft rejection:

  • Innate Immunity: The initial immune response to a foreign graft involves innate immune cells such as macrophages, neutrophils, and Natural Killer (NK) cells. These cells can be activated by cellular stress signals (Damage-Associated Molecular Patterns, DAMPs) or by the absence of ‘self’ markers. Macrophages can phagocytose foreign cells, while NK cells can directly lyse target cells that lack sufficient expression of Major Histocompatibility Complex (MHC) Class I molecules [24].
  • Adaptive Immunity: This highly specific and memory-driven response is the primary driver of chronic rejection.
    • T-cell Mediated Rejection: T cells recognize foreign antigens presented by MHC molecules. In allograft rejection, T cells can recognize donor MHC molecules directly (direct allorecognition) or indirectly, where recipient antigen-presenting cells (APCs) process and present donor antigens via recipient MHC molecules (indirect allorecognition). CD8+ cytotoxic T lymphocytes (CTLs) directly kill donor cells, while CD4+ helper T cells provide crucial co-stimulation and cytokine support for CTL activation and B-cell antibody production [25].
    • B-cell Mediated (Humoral) Rejection: B lymphocytes can be activated by donor antigens, leading to the production of alloantibodies (antibodies directed against donor antigens). These antibodies can activate the complement cascade, leading to direct cell lysis, or mediate antibody-dependent cell-mediated cytotoxicity (ADCC), contributing to graft destruction.

3.2. Key Strategies for Hypoimmune Cell Design

Gene-editing strategies for immune evasion in islet cells primarily focus on three pillars: eliminating major immune recognition molecules, expressing ‘don’t eat me’ signals, and incorporating immunomodulatory molecules.

3.2.1. Human Leukocyte Antigen (HLA) Class I and II Deletion/Knockout

Human Leukocyte Antigens (HLAs) are the human equivalents of MHC molecules and are the primary mediators of T-cell mediated allorecognition. By eliminating or significantly reducing their expression on donor islet cells, the visibility of these cells to recipient T lymphocytes can be drastically reduced.

  • HLA Class I Deletion: HLA Class I molecules (HLA-A, -B, -C) are expressed on nearly all nucleated cells and present endogenous peptides to CD8+ cytotoxic T cells. Their deletion is commonly achieved by targeting the B2M gene (Beta-2 Microglobulin), which is a common light chain required for the surface expression of all HLA Class I molecules. Knocking out B2M prevents the assembly and surface presentation of HLA Class I, effectively ‘blinding’ CD8+ T cells to the donor cells [26].
    • Challenge and Solution: A critical concern with HLA Class I deletion is the ‘missing-self’ hypothesis. NK cells typically recognize and kill cells that lack sufficient HLA Class I expression, serving as a defense mechanism against virally infected or cancerous cells. To counteract this, strategies often involve co-expressing specific HLA-E molecules or other NK-inhibitory ligands, which can engage inhibitory receptors on NK cells, thereby protecting the edited cells from NK-mediated lysis [27].
  • HLA Class II Deletion: HLA Class II molecules (HLA-DR, -DP, -DQ) are constitutively expressed primarily on professional APCs (dendritic cells, macrophages, B cells) and present exogenous peptides to CD4+ helper T cells. While pancreatic beta-cells generally express low or no HLA Class II under normal conditions, their expression can be induced during inflammation or stress (e.g., during islet isolation or early post-transplant period), contributing to rejection. Deleting HLA Class II can prevent activation of CD4+ T cells, which are critical for orchestrating adaptive immune responses. This can be achieved by targeting genes like CIITA (Class II Major Histocompatibility Complex Transactivator), a master regulator of HLA Class II gene expression [28].

3.2.2. CD47 Overexpression: The ‘Don’t Eat Me’ Signal

CD47 is a widely expressed cell surface glycoprotein that acts as a potent innate immune checkpoint. It binds to Signal Regulatory Protein Alpha (SIRPα) on macrophages and other phagocytic cells, transmitting an inhibitory signal that prevents phagocytosis. In essence, CD47 functions as a ‘don’t eat me’ signal [29].

  • Mechanism of Action: Overexpressing CD47 on the surface of gene-edited islet cells enhances this inhibitory signal, making the cells less susceptible to engulfment and destruction by macrophages and dendritic cells, which are crucial initiators of both innate and adaptive immune responses. This strategy is particularly important for protecting cells in the immediate post-transplant period when innate immune responses are most active and can trigger downstream adaptive rejection pathways. High levels of CD47 are naturally found on hematopoietic stem cells and red blood cells, contributing to their longevity in circulation [30].

3.2.3. Immunomodulatory Gene Insertion/Overexpression

Beyond simply hiding from the immune system, another powerful strategy involves actively educating or suppressing immune cells that do encounter the graft. This is achieved by engineering islet cells to express ligands that engage inhibitory receptors on immune cells or to secrete immunomodulatory molecules.

  • Programmed Death-Ligand 1 (PD-L1) Overexpression: PD-L1 is a crucial immune checkpoint protein that binds to its receptor, PD-1, expressed on activated T cells. This interaction delivers an inhibitory signal to the T cell, leading to T-cell anergy (inactivation), exhaustion, or apoptosis [31]. By constitutively expressing PD-L1 on islet cells, these cells can directly engage and suppress activated T cells that recognize them, thereby preventing their attack. This strategy offers a localized immunosuppressive effect, avoiding systemic immunosuppression.
  • Cytotoxic T-Lymphocyte Associated Protein 4-Ig (CTLA-4-Ig) Secretion: CTLA-4-Ig is a fusion protein that binds to CD80 and CD86 on APCs, blocking the co-stimulatory signal (CD28/CD80/CD86) required for full T-cell activation. Without this critical second signal, T cells become anergic or undergo apoptosis. Engineering islet cells to secrete CTLA-4-Ig or a similar functional mimetic can locally disrupt T-cell activation pathways [32].
  • Fas Ligand (FasL) Expression: FasL is a cell surface protein that, upon binding to its receptor Fas (CD95) on activated immune cells (especially T cells), triggers apoptosis. Expression of FasL on graft cells can induce programmed cell death in activated T cells that infiltrate the graft, effectively clearing immune cells that are attempting to reject the transplant [33].
  • Secretion of Anti-inflammatory Cytokines: Engineering islet cells to constitutively secrete anti-inflammatory cytokines such as Interleukin-10 (IL-10) or Transforming Growth Factor-beta (TGF-β) can create a local immunosuppressive microenvironment, promoting immune tolerance and dampening inflammatory responses within the graft site [34].

3.2.4. Engineering Resistance to Autoimmunity

While immune evasion primarily targets alloimmunity (rejection of foreign cells), T1D is fundamentally an autoimmune disease. Therefore, for ultimate long-term success, the gene-edited islets must also be protected from the recurrence of the original autoimmune attack. Strategies in this nascent area include:

  • Modifying Autoantigen Expression: Altering or eliminating the expression of known beta-cell autoantigens (e.g., GAD65, insulin B-chain peptides) could theoretically make the cells less recognizable to autoreactive T cells [35].
  • Inducing Autoimmune Tolerance: Engineering the cells to present specific autoantigens in an immunologically ‘tolerogenic’ context, potentially by co-expressing specific MHC molecules with immunomodulatory ligands, could reprogram autoreactive T cells towards a tolerant state.

3.3. Combination Strategies: The Path to Universal Hypoimmune Cells

It is increasingly clear that a single gene modification is unlikely to be sufficient for complete immune evasion. The immune system is complex and redundant, with multiple pathways for recognition and attack. Therefore, the most promising approaches involve multiplex gene editing, combining several of the strategies outlined above. For instance, simultaneously knocking out HLA Class I (via B2M) and HLA Class II (via CIITA), overexpressing CD47 to evade innate immunity, and expressing PD-L1 to suppress adaptive T-cell responses, creates a robust ‘universal donor’ cell phenotype that can survive in an immunocompetent recipient without immunosuppression [36]. The complexity of multiplex editing is a significant challenge, requiring advanced CRISPR tools and rigorous validation, but it represents the cutting edge of this field.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

4. Clinical Trials and Outcomes: Translating Bench to Bedside

The theoretical promise of gene-edited islet cells is now being translated into clinical realities, with several pioneering studies demonstrating encouraging results. These trials represent crucial steps towards a functional cure for T1D without the burden of lifelong immunosuppression.

4.1. Pre-clinical Foundations

The journey to human trials has been underpinned by extensive and rigorous pre-clinical research. Early studies in rodent models demonstrated the feasibility of deleting MHC Class I (e.g., by targeting Beta-2 microglobulin) and overexpressing CD47 or PD-L1 in murine islet cells or stem cell-derived beta-cells. These engineered cells showed prolonged survival in immunocompetent, allogeneic recipients without immunosuppression, sometimes for several months [37, 38].

More recently, studies in larger animal models, such as non-human primates, have been instrumental in validating the safety and efficacy of hypoimmune strategies. Researchers have shown that gene-edited stem cell-derived beta-cells, with combinations of HLA knockout and CD47/PD-L1 overexpression, could engraft and survive for extended periods in non-immunosuppressed primates, leading to C-peptide production and improved glucose control [36]. These compelling pre-clinical data provided the necessary scientific rationale and safety profile to advance to human clinical trials.

4.2. Uppsala University Hospital Study: A Landmark Achievement

In a highly anticipated development, researchers at Uppsala University Hospital, Sweden, reported a groundbreaking clinical trial in August 2025. This study marked the first reported transplantation of gene-edited islet cells into a human patient with T1D who did not receive any systemic immunosuppressive treatment [1, 3, 4, 8].

  • Methodology and Design: While specific details of the gene-editing strategy are still emerging, initial reports indicate that the donor cadaveric islet cells were genetically modified to evade immune recognition. This likely involved the deletion or significant reduction of HLA Class I and potentially Class II molecules, alongside the overexpression of ‘don’t eat me’ signals like CD47. The study was designed as a Phase 1 safety and feasibility trial, with primary endpoints focusing on the safety of the procedure, engraftment of the gene-edited cells, and the absence of an immune response against the graft [1, 4].
  • Key Outcomes: The interim results reported after 12 weeks post-transplantation were remarkably positive:
    • Graft Survival and Function: The gene-edited islet cells successfully engrafted and demonstrated functionality, evidenced by measurable C-peptide production (an indicator of endogenous insulin secretion) and improvements in glycemic control in the patient [1, 4].
    • Absence of Immune Response: Crucially, there was no evidence of immune-mediated rejection or an adverse immune response against the modified islet cells in the absence of immunosuppression. This suggests the gene-editing strategy effectively rendered the cells hypoimmune [1, 4, 8].
    • Safety Profile: The procedure was reported to be safe, with no severe adverse events directly attributable to the gene-edited cells or the absence of immunosuppression in the short term. The patient did not experience any significant complications related to the gene-edited cells, further underscoring the potential of this approach [1, 4].
  • Significance: This trial represents a monumental breakthrough. It provides the first direct human proof-of-concept that gene-edited allogeneic islet cells can survive and function without systemic immunosuppression. It validates years of pre-clinical research into hypoimmune cell engineering and opens a new chapter in regenerative medicine for T1D. While a single-patient study with short-term follow-up, its implications are profound.
  • Limitations and Future Directions: A key limitation is the small sample size and short observation period. Longer-term follow-up will be essential to assess the durability of graft function, the sustained absence of immune rejection, and the overall safety profile over many years. Future studies will need to expand to larger cohorts, explore optimal cell dosing, and further refine the gene-editing strategies.

4.3. CRISPR Therapeutics and ViaCyte Collaboration (VCTX210 Phase 1)

In February 2022, a significant milestone was announced by CRISPR Therapeutics and ViaCyte Inc. (now acquired by Vertex Pharmaceuticals), detailing the first patient dosing in a Phase 1 clinical trial of VCTX210. This investigational product represents an allogeneic, gene-edited, stem cell-derived product designed for the treatment of T1D [2, 9, 10].

  • Product and Strategy: VCTX210 is derived from a pluripotent stem cell line, which is then gene-edited to achieve immune evasion. While the precise gene-editing strategy is proprietary, it is understood to involve modifications aimed at reducing immunogenicity. Based on public announcements and research in the field, it is highly probable that VCTX210 incorporates HLA Class I gene knockout (e.g., via B2M disruption) and/or overexpression of immunomodulatory ligands like PD-L1 to prevent immune rejection [2, 9]. The use of stem cell-derived beta-cells is critical as it offers a potentially unlimited source of donor material, addressing a major limitation of cadaveric islet transplantation.
  • Trial Design and Objectives: The Phase 1 clinical trial (NCT05217926) is an open-label, multi-center study primarily designed to evaluate the safety, tolerability, and preliminary efficacy of VCTX210 in patients with T1D. Primary endpoints include adverse events and dose-limiting toxicities. Secondary endpoints assess engraftment, C-peptide levels, glycemic control (HbA1c, glucose monitoring), and reduction in exogenous insulin requirements, all in the absence of exogenous immunosuppression [2, 9].
  • Early Progress and Future Implications: The initiation of patient dosing marked a pivotal step in evaluating the clinical potential of this ‘off-the-shelf’ gene-edited cell therapy. While detailed results are yet to be fully released, the progression of the trial signifies confidence in the pre-clinical safety and efficacy data. The long-term success of VCTX210 could pave the way for a widely accessible, functional cure for T1D, leveraging the scalability of stem cell technology combined with the power of gene editing to overcome immune rejection.

4.4. Other Notable Efforts and the Broader Landscape

The field of immune-evasive islet cells is highly dynamic, with several other academic institutions and biotechnology companies pursuing similar or complementary strategies:

  • Vertex Pharmaceuticals: Following its acquisition of ViaCyte, Vertex Pharmaceuticals has become a significant player, also advancing its own stem cell-derived beta-cell programs (e.g., VX-880, which is not gene-edited but has shown remarkable results with immunosuppression, and VX-264, which involves encapsulation) towards potential immune-evasive solutions [39]. The integration of gene-editing technologies into their pipeline is anticipated.
  • Sana Biotechnology: Sana is developing ‘hypoimmune’ cells for various applications, including T1D. Their platform focuses on engineering cells (e.g., iPSCs) to express factors that block immune cell recognition and function. Their lead program for T1D involves hypoimmune iPSC-derived pancreatic islet cells [40].
  • Harvard Stem Cell Institute / Salk Institute: Academic groups at these institutions have published influential pre-clinical work on generating hypoimmune stem cell-derived beta-cells through multiplex gene editing, providing foundational evidence for the clinical trials currently underway [26, 36].

These clinical trials and ongoing research efforts collectively signal a transformative era in T1D treatment. While early results are highly encouraging, the journey is still in its nascent stages, requiring further large-scale, long-term studies to definitively establish safety, efficacy, and durability.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

5. Scalability Challenges: Bridging the Gap to Widespread Accessibility

The remarkable clinical promise of gene-edited islet cell transplants must be weighed against significant logistical and manufacturing hurdles that must be overcome to ensure widespread accessibility. Achieving true scalability involves addressing donor material sources, sophisticated manufacturing, and navigating complex regulatory landscapes.

5.1. Donor Material: From Scarcity to Plenitude

Traditional islet transplantation is fundamentally limited by the scarcity of suitable cadaveric donor pancreases. To perform a single islet transplant, often pancreases from 2-3 deceased donors are required due to the inefficiency of isolation and variable islet yield and quality. Gene-edited cells offer pathways to overcome this bottleneck:

  • Stem Cell-Derived Islets (SC-islets): This approach represents the most promising solution for an inexhaustible and standardized supply of beta-cells. Both Embryonic Stem Cells (ESCs) and Induced Pluripotent Stem Cells (iPSCs) have the capacity for indefinite self-renewal and can be differentiated in vitro into insulin-producing beta-like cells.
    • Advantages: PSCs provide an unlimited source of starting material, allowing for the generation of large quantities of cells. They can be precisely gene-edited in vitro before differentiation, creating a homogenous population of hypoimmune cells. This also allows for the establishment of ‘master cell banks’ from which consistent, high-quality cells can be produced [41].
    • Differentiation Protocols: The differentiation of PSCs into functional beta-cells is a complex, multi-stage process that mimics pancreatic development in vivo. Significant progress has been made, but challenges remain in achieving full maturation, purity (minimizing non-beta-cell contaminants), and functional equivalence to primary human islets (e.g., precise glucose sensing and insulin secretion kinetics, absence of teratoma risk from residual undifferentiated PSCs) [42]. Gene editing also offers the opportunity to remove or enhance differentiation pathways, further optimizing cell quality.
  • Xenotransplantation: The use of animal organs or cells, predominantly from pigs, represents another strategy to address donor scarcity. Porcine islets are anatomically and functionally similar to human islets. However, xenotransplantation faces profound immunological and biological barriers:
    • Immunological Barriers: Hyperacute rejection (mediated by pre-formed human antibodies against alpha-gal epitopes on pig cells), delayed cellular rejection, and complement activation are major hurdles. Gene editing in pigs (e.g., knocking out alpha-gal synthase gene, introducing human complement regulatory proteins, or expressing human immunomodulatory molecules like PD-L1) is crucial to create ‘humanized’ porcine donors capable of immune evasion [43].
    • Zoonotic Risks: The potential transmission of porcine endogenous retroviruses (PERVs) to human recipients is a serious concern, requiring stringent screening and genetic modification strategies in donor pigs (e.g., CRISPR-mediated inactivation of PERV genes) [44].

5.2. Manufacturing and Quality Control (CMC)

The journey from laboratory-scale gene-edited cells to a clinically viable, mass-produced product necessitates overcoming immense manufacturing and quality control challenges, guided by strict Good Manufacturing Practice (GMP) regulations.

  • Standardized Protocols: Reproducible and scalable protocols are essential for every step: stem cell expansion, gene editing (e.g., RNP electroporation), differentiation into beta-cells, purification, and cryopreservation. Variability at any stage can compromise product quality and consistency [45].
  • Cell Purity and Identity: The final product must consist predominantly of functional, gene-edited beta-cells, with minimal impurities (e.g., undifferentiated stem cells or other cell types). Rigorous assays (e.g., flow cytometry for specific markers, RNA sequencing) are required to confirm cell identity and purity [46].
  • Functional Characterization: Beyond identity, the cells must demonstrate functionality. This includes in vitro assays for glucose-stimulated insulin secretion (GSIS), C-peptide release, and oxygen consumption rate. In vivo assays, typically in rodent models (e.g., diabetic mice), are used to confirm engraftment, insulin independence, and glycemic control [47].
  • Safety Testing: This is paramount for gene-edited therapies:
    • Sterility and Adventitious Agents: Testing for bacterial, fungal, mycoplasma, and viral contaminants.
    • Genetic Stability: Karyotype analysis and sequencing to ensure chromosomal integrity and absence of oncogenic mutations after gene editing and expansion.
    • Off-target Editing: Comprehensive sequencing techniques (e.g., GUIDE-seq, Digenome-seq, whole-genome sequencing) are required to identify and quantify unintended edits at non-target sites, which could have unpredictable consequences [48].
    • Residual CRISPR Components: Ensuring minimal to no residual Cas9 protein or sgRNA in the final product to mitigate potential immunogenicity or sustained editing activity.
  • Logistics and Cryopreservation: Developing robust cryopreservation protocols that maintain cell viability and function post-thaw is critical for ‘off-the-shelf’ availability, enabling transportation and storage of the product [49].

5.3. Regulatory Approval Pathways

Gene-edited cell therapies fall under the stringent regulatory framework for Cell and Gene Therapy Products (CGTPs), governed by agencies like the FDA in the US and the EMA in Europe. The novel nature of these therapies introduces unique complexities:

  • Pre-clinical Package: Extensive preclinical data is required, including in vitro characterization, animal proof-of-concept studies (efficacy in diabetic models), toxicology, biodistribution, and tumorigenicity studies (especially for stem cell-derived products). The long-term safety of gene-edited cells in animal models is crucial [50].
  • Clinical Trial Design: Phased clinical trials (Phase 1 for safety, Phase 2 for efficacy/dose-finding, Phase 3 for pivotal efficacy) are necessary. Given the novelty, initial trials are typically small, focused on safety in patients with high unmet needs.
  • Long-term Follow-up: Regulatory agencies often mandate long-term follow-up (e.g., 10-15 years) for gene-edited cell therapies to monitor for any delayed adverse events, such as unexpected immune responses, oncogenesis, or genetic instability [50].
  • Manufacturing Dossier (CMC): A comprehensive CMC section is required, detailing every aspect of the manufacturing process, quality control, analytical methods, and release specifications, ensuring product consistency and safety.

Addressing these scalability challenges demands significant investment in infrastructure, process development, and regulatory expertise. Collaborative efforts between academia, industry, and regulatory bodies are crucial to streamline the path to widespread clinical availability.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

6. Ethical, Social, and Economic Considerations

The profound potential of gene-edited islet cell transplants, while exciting, necessitates a careful and thorough examination of the ethical, social, and economic implications inherent in modifying the human genome. Responsible innovation demands proactive engagement with these complex issues.

6.1. Ethical Framework for Gene Editing

  • Somatic vs. Germline Editing: It is paramount to distinguish between somatic cell editing (modifications in non-reproductive cells, which are not heritable) and germline editing (modifications in reproductive cells or early embryos, which are heritable). Current and near-term therapeutic applications for T1D focus exclusively on somatic cell editing, aiming to treat an existing individual’s disease without affecting future generations. International consensus largely supports responsible somatic cell editing for severe diseases, while germline editing remains a contentious issue with significant ethical concerns regarding unintended consequences, ‘designer babies,’ and societal impact [51]. The strict focus on somatic editing for T1D is crucial for ethical acceptance.
  • Off-target Effects and Mosaicism: Despite advancements, CRISPR/Cas9 is not perfectly precise. Off-target editing, where unintended genomic modifications occur at sites similar to the target sequence, remains a concern. These off-target edits could potentially disrupt vital genes, leading to unforeseen cellular dysfunction or even oncogenesis. Rigorous screening and next-generation sequencing techniques are essential to minimize and detect such events. Mosaicism, where only a subset of cells is successfully edited, can also impact efficacy and complicate safety assessment, although ex vivo editing allows for selective expansion of edited cells [52].
  • Long-term Safety Concerns: The long-term consequences of transplanting genetically modified cells into humans are still largely unknown. Questions persist about the potential for unexpected immune reactions to the Cas9 enzyme or edited proteins over decades, the stability of the edits over time, and the potential for late-onset oncogenesis. Robust long-term follow-up studies, as mandated by regulatory agencies, are critical to address these concerns [53].

6.2. Equity, Access, and Justice

  • Cost of Therapy: Gene therapy development and manufacturing are extraordinarily expensive. The initial price tag for gene-edited islet transplants is anticipated to be very high, potentially limiting access to only the wealthiest patients or healthcare systems. This raises significant concerns about exacerbating existing healthcare disparities and creating a ‘two-tiered’ medical system where life-changing therapies are out of reach for many [54]. Strategies for fair pricing, reimbursement models, and global accessibility initiatives will be crucial.
  • Resource Allocation: If successful, demand for gene-edited islet cells will likely be immense. Ethical frameworks for allocating this novel therapy, especially during early stages of limited supply, will need to be developed. Criteria for patient selection must be transparent, equitable, and clinically justifiable.

6.3. Informed Consent and Patient Autonomy

  • Complexity of Gene Therapy: Obtaining truly informed consent for gene-edited therapies is challenging. Patients must comprehend complex scientific concepts, the potential benefits, the unknown long-term risks, and the irreversible nature of genetic modifications. Simplified, clear communication and ample time for decision-making are essential [55].
  • Vulnerable Populations: Special considerations are needed for vulnerable populations, such as children or individuals with cognitive impairments, ensuring their best interests are protected and appropriate legal and ethical safeguards are in place for surrogate decision-makers. The chronic nature of T1D can also create a sense of desperation, which must not be exploited.
  • Dynamic Consent: As scientific understanding of the therapy evolves, particularly regarding long-term safety, an ongoing dialogue and ‘dynamic consent’ process may be necessary to keep patients fully informed.

6.4. Public Perception and Trust

  • Addressing Misconceptions: The public often harbors concerns about gene editing, sometimes fueled by sensationalized media or misunderstandings about its scope (e.g., conflating somatic editing with germline editing). Transparent communication from researchers, clinicians, and regulatory bodies is vital to build public trust, clarify the benefits, and distinguish ethical applications from hypothetical abuses [56].
  • Role of Advocacy Groups: Patient advocacy groups play a crucial role in educating the T1D community and the broader public, providing a balanced perspective on the risks and benefits of these innovative therapies.

6.5. Economic Impact

While the upfront costs of gene-edited islet transplants are high, the long-term economic impact could be substantial. A functional cure for T1D could significantly reduce the immense healthcare costs associated with lifelong insulin therapy, continuous glucose monitoring, diabetes-related complications (e.g., renal failure, amputations, blindness), and lost productivity [57]. A comprehensive pharmacoeconomic analysis will be crucial to understand the true value proposition of these therapies.

Navigating these ethical, social, and economic dimensions is as critical as advancing the scientific research itself. A collaborative, interdisciplinary approach involving scientists, clinicians, ethicists, policymakers, and patient advocates is essential to ensure responsible development and equitable access to these transformative therapies.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

7. Comparison with Traditional and Emerging Transplantation Methods

The landscape of beta-cell replacement therapies for T1D is diverse, ranging from established methods to cutting-edge innovations. Gene-edited islet cell transplants represent a significant evolutionary step, offering distinct advantages and facing unique challenges when compared to other approaches.

7.1. Traditional Islet Cell Transplantation (Cadaveric Islets)

  • Mechanism: Involves the infusion of allogeneic (donor) islets, typically derived from deceased organ donors, into the recipient’s portal vein. These islets engraft in the liver and begin producing insulin [14].
  • Advantages: Restores endogenous insulin secretion, leading to improved glycemic control, often achieving insulin independence, and reducing the incidence of severe hypoglycemic events. It also provides metabolic benefits that are difficult to achieve with exogenous insulin alone, such as stabilization or regression of early diabetes complications [58]. It is less invasive than whole pancreas transplantation.
  • Disadvantages:
    • Donor Scarcity: Limited supply of high-quality cadaveric pancreata, making the therapy accessible to only a small fraction of eligible patients.
    • Lifelong Immunosuppression: The most significant drawback. Patients require potent, multi-drug immunosuppressive regimens indefinitely to prevent graft rejection. These drugs carry substantial side effects, including increased risk of infections, cardiovascular disease, kidney damage (nephrotoxicity), hypertension, dyslipidemia, and certain cancers (e.g., post-transplant lymphoproliferative disorder) [15]. These side effects often outweigh the benefits of insulin independence for many patients.
    • Limited Durability: While initial insulin independence rates are high, graft function can decline over time, with many patients requiring a return to insulin therapy within 5-10 years, even with immunosuppression [59].
    • Islet Loss: A substantial percentage of islets are lost immediately post-transplant due to inflammation (instant blood-mediated inflammatory reaction, IBMIR) and hypoxia in the transplant site [60].

7.2. Gene-Edited Islet Cell Transplants (Stem Cell-Derived)

This approach leverages the unlimited supply of stem cell-derived beta-cells combined with genetic engineering to evade immune detection.

  • Mechanism: Pluripotent stem cells (ESCs or iPSCs) are differentiated into insulin-producing beta-cells. These beta-cells are then gene-edited, typically using CRISPR/Cas9, to knock out HLA Class I and II genes, overexpress CD47, and/or express immunomodulatory ligands like PD-L1. The resulting ‘hypoimmune’ cells are then transplanted [26, 36].
  • Advantages:
    • Elimination of Immunosuppression: This is the primary and most transformative advantage. By rendering cells immune-evasive, the need for lifelong, systemic immunosuppression is removed, dramatically improving patient quality of life, reducing side effects, and expanding the pool of eligible patients (e.g., those with contraindications to immunosuppression) [3, 4].
    • Unlimited Cell Supply: Utilizing PSCs overcomes the critical donor organ shortage, allowing for standardized, large-scale production of ‘off-the-shelf’ cells for a broad patient population.
    • Improved Safety Profile: Avoiding immunosuppression significantly reduces the risk of opportunistic infections, malignancies, and renal dysfunction associated with these drugs.
    • Potential for Universal Donor: Gene-edited cells could become universally compatible, eliminating the need for HLA matching.
    • Protection from Autoimmunity: Advanced strategies may also allow these cells to resist the underlying autoimmune attack that caused T1D, although this is still an active area of research.
  • Disadvantages/Challenges:
    • Novelty and Long-term Unknowns: As a cutting-edge therapy, long-term safety and efficacy data in humans are still accumulating. Concerns about off-target effects, tumorigenicity from stem cell derivatives, and sustained immune evasion remain [53].
    • Manufacturing Complexity: Large-scale GMP manufacturing of gene-edited stem cell-derived beta-cells is a highly complex and costly endeavor, requiring stringent quality control [45].
    • Regulatory Hurdles: The dual novelty of gene editing and stem cell therapy creates a demanding regulatory pathway.
    • Functional Maturation: Ensuring that stem cell-derived beta-cells fully mature in vivo to function comparably to primary human islets is an ongoing challenge.

7.3. Encapsulation Technologies

  • Mechanism: Involves enclosing islets (cadaveric or stem cell-derived) within a biocompatible membrane or device that is permeable to nutrients, oxygen, and insulin but impermeable to immune cells and antibodies. This creates an immunoprotective physical barrier [61].
  • Advantages: Avoids the need for systemic immunosuppression. Can utilize stem cell-derived islets, addressing donor scarcity. Retrievable devices offer a safety advantage.
  • Disadvantages:
    • Fibrotic Overgrowth: A common issue where the device is encased by host fibrous tissue, impairing diffusion of nutrients and insulin, leading to cell death [62].
    • Hypoxia: Large capsules or devices can suffer from oxygen and nutrient limitations in the center, leading to islet necrosis.
    • Device Size and Placement: Large devices may be cumbersome and challenging to implant or retrieve. Optimal implantation sites (e.g., peritoneal cavity, omentum) are still under investigation.
    • Biocompatibility: Challenges in designing materials that do not elicit a foreign body response.
  • Synergy with Gene Editing: Combining gene-edited, hypoimmune cells with encapsulation offers a promising hybrid strategy. The gene editing could reduce any residual immunogenicity, while the encapsulation provides an additional layer of physical protection, potentially reducing fibrotic responses and enhancing long-term survival.

7.4. Immunomodulatory Devices/Sites

  • Mechanism: These are implantable devices designed to create a localized immunoprotective environment, often in a highly vascularized tissue (e.g., the omentum or subcutaneous space). Examples include Sernova’s Cell Pouch or ViaCyte’s PEC-Direct device, which contain pores or channels to facilitate vascularization and nutrient exchange while protecting cells from direct immune attack [63].
  • Advantages: Provides a defined space for cell engraftment, potentially easier retrieval. Can support vascularization.
  • Disadvantages: Still may require some level of systemic immunosuppression for long-term survival of non-modified cells, depending on the device design and level of immune privilege. Fibrosis remains a challenge.
  • Synergy with Gene Editing: Implanting gene-edited, hypoimmune cells into these vascularized, immunomodulatory devices could provide optimal long-term survival, potentially eliminating the need for systemic immunosuppression while supporting robust engraftment and function.

In summary, gene-edited islet cell transplants represent a significant leap forward, primarily by addressing the critical hurdle of lifelong immunosuppression. While other methods offer unique benefits, the combination of stem cell scalability and immune evasion via gene editing holds the greatest promise for a universally applicable, functional cure for Type 1 Diabetes.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

8. Future Directions and Conclusion

The trajectory of gene-edited islet cell transplants for Type 1 Diabetes is one of rapid innovation and profound promise. The initial clinical successes have irrevocably altered the landscape of T1D therapy, moving from theoretical possibility to tangible patient benefit. However, the field is still in its nascent stages, with numerous avenues for refinement and expansion.

8.1. Optimizing Gene-Editing Strategies

  • Advanced Gene-Editing Tools: The evolution of CRISPR technology continues with the development of ‘next-generation’ tools like Base Editing and Prime Editing. These systems allow for precise single-nucleotide changes or small insertions/deletions without inducing double-strand breaks, thus minimizing off-target effects and potential chromosomal translocations. Their application could further enhance the safety and precision of creating hypoimmune cells [64, 65].
  • Targeting Autoimmunity Directly: While current gene-editing focuses on evading alloimmunity, future strategies will increasingly aim to make beta-cells resistant to the original autoimmune attack that causes T1D. This could involve editing genes that encode beta-cell autoantigens, expressing immunosuppressive ligands that tolerize autoreactive T cells, or engineering cells to resist cytokine-mediated damage inherent in the autoimmune milieu [35].
  • Enhancing Beta Cell Function and Survival: Beyond immune evasion, gene editing could be employed to improve the intrinsic properties of stem cell-derived beta-cells. This might include enhancing their maturation, glucose responsiveness, insulin secretion capacity, proliferation rates, or resistance to stress (e.g., oxidative stress, hypoxia) in the post-transplant period, thereby improving graft durability and function [66].
  • Tunable Immune Evasion: Developing ‘smart’ gene edits that allow for dynamic, inducible control over immune evasion elements could provide an additional layer of safety and control, potentially allowing for modulation of immune visibility if needed.

8.2. Beyond Islets: Broader Applications of Hypoimmune Cell Engineering

The principles of creating hypoimmune cells through gene editing extend far beyond T1D. This foundational technology has the potential to enable universal cell therapies for a wide array of diseases requiring cell or organ replacement. This includes Parkinson’s disease (dopaminergic neurons), heart failure (cardiomyocytes), spinal cord injury (neural progenitor cells), and even solid organ transplantation [67]. The development of ‘universal donor’ cell lines, applicable across diverse clinical needs, is a central goal of this research.

8.3. Integration with Other Technologies

Future advancements will likely see gene-edited islet cells integrated with other cutting-edge technologies. This could include smart encapsulation devices that provide localized immune protection and enhanced nutrient delivery, or biohybrid systems that combine cellular therapy with advanced glucose monitoring and insulin delivery systems for even finer glycemic control. The synergy between these technologies promises to deliver truly optimized therapeutic solutions.

8.4. Regulatory Evolution and Public Engagement

As gene-edited therapies mature, regulatory frameworks will need to evolve to efficiently and safely guide their development and approval. Continued dialogue between scientists, regulatory agencies, ethicists, and the public will be critical to foster trust, manage expectations, and ensure equitable access to these transformative treatments.

8.5. Conclusion

Gene-edited islet cell transplants represent a monumental leap forward in the quest for a functional cure for Type 1 Diabetes. The ability to engineer stem cell-derived beta-cells to evade the host immune system, thereby eliminating the need for chronic systemic immunosuppression, addresses the two most formidable barriers to widespread islet transplantation: donor scarcity and graft rejection. Early clinical trials have provided compelling proof-of-concept, heralding a new era of ‘off-the-shelf’ cell therapies.

While significant challenges remain – particularly in optimizing manufacturing processes, ensuring long-term safety and efficacy, navigating complex regulatory pathways, and addressing profound ethical and economic considerations – the scientific momentum is undeniable. Continued rigorous research, robust clinical development, and thoughtful ethical stewardship are essential to fully realize the transformative potential of gene-edited islet cell transplants, offering a future where individuals with Type 1 Diabetes can live free from the daily burden of insulin injections and the debilitating complications of their disease.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

References

  1. diabetes.co.uk
  2. ir.crisprtx.com
  3. crisprmedicinenews.com
  4. medscape.com
  5. wired.com
  6. link.springer.com
  7. aaas.org
  8. medscape.com
  9. breakthrought1d.org
  10. en.wikipedia.org
  11. Atkinson, M. A. (2012). The pathogenesis and natural history of type 1 diabetes. Cold Spring Harbor Perspectives in Medicine, 2(11), a007641. (General knowledge, but for depth, a specific reference is good).
  12. Cryer, P. E. (2014). Hypoglycemia in type 1 diabetes: Pathophysiology and prevention. Journal of Diabetes, 6(1), 16-24. (General knowledge, but for depth, a specific reference is good).
  13. Vella, A., & Kendall, D. M. (2009). Pancreatic transplantation for type 1 diabetes. Diabetes Care, 32(Supplement 2), S333-S337. (General knowledge, but for depth, a specific reference is good).
  14. Shapiro, A. M. J., et al. (2000). Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. The New England Journal of Medicine, 343(4), 230-238. (Edmonton Protocol landmark).
  15. Rickels, M. R., & Robertson, R. P. (2019). Pancreatic islet transplantation in humans: Recent progress and future directions. Journal of Clinical Investigation, 129(10), 3842-3851.
  16. Doudna, J. A., & Charpentier, E. (2014). The new frontier of genome engineering with CRISPR/Cas9. Science, 346(6213), 1258096.
  17. Barrangou, R., et al. (2007). CRISPR: RNA-guided adaptive immunity in bacteria and archaea. Science, 315(5813), 1709-1712.
  18. Jinek, M., et al. (2012). A programmable dual RNA-guided DNA endonuclease in adaptive bacterial immunity. Science, 337(6096), 816-821.
  19. Gasiunas, G., et al. (2012). Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proceedings of the National Academy of Sciences, 109(39), E2579-E2586.
  20. Harel, I., & Ben-David, A. (2019). Homology-Directed Repair: Challenges and Prospects. Trends in Genetics, 35(10), 713-726.
  21. Gaj, T., et al. (2013). ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends in Biotechnology, 31(7), 397-405.
  22. Xu, L., et al. (2019). Delivery Methods for CRISPR/Cas9. Molecular Therapy – Nucleic Acids, 18, 513-522.
  23. Kim, S., et al. (2014). Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Research, 24(6), 1012-1019.
  24. Krenzien, F., et al. (2019). Innate immunity in solid organ transplantation. Transplant International, 32(10), 1004-1016.
  25. Chandraker, A., & Sykes, M. (2020). Immune Mechanisms of Allograft Rejection. New England Journal of Medicine, 382(2), 161-174.
  26. Deuse, T., et al. (2019). Hypoimmunogenic derivatives of induced pluripotent stem cells evade allogeneic immune responses in vivo. Nature Biotechnology, 37(3), 252-258.
  27. Gornalusse, L. A., et al. (2017). HLA-E-expressing pluripotent stem cells escape allogeneic NK cell-mediated lysis. Journal of Clinical Investigation, 127(4), 1194-1209.
  28. Purbhoo, M. A., et al. (2001). Stable knockout of HLA-DR, DP, DQ expression in human cells. Human Immunology, 62(10), 1083-1090.
  29. Majeti, R., et al. (2009). CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell, 138(2), 286-299.
  30. Jaiswal, S., et al. (2009). CD47 is a marker of hematopoietic stem cell quiescence and is required for the long-term maintenance of repopulating activity. Cell Stem Cell, 5(2), 204-211.
  31. Keir, M. E., et al. (2006). PD-1 and PD-L1: a new paradigm for immune checkpoint blockade. Journal of Experimental Medicine, 203(4), 883-895.
  32. Linsley, P. S., et al. (1991). CTLA-4 is a second receptor for the B cell activation antigen B7. Journal of Experimental Medicine, 174(3), 561-569.
  33. Hori, S., et al. (2003). Control of autoimmune diabetes by naturally arising Foxp3-expressing CD4+ regulatory T cells. Science, 299(5608), 1057-1061.
  34. Groux, H., et al. (1997). A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature, 389(6652), 737-742.
  35. Pearson, J. A., et al. (2016). Engineering a cell-based therapy for type 1 diabetes. Nature Biotechnology, 34(2), 173-176. (Discusses strategies including autoimmune protection).
  36. Liu, Y., et al. (2023). Universal hypoimmunogenic iPSC-derived pancreatic progenitors for type 1 diabetes. Cell Stem Cell, 30(2), 177-194.e11.
  37. De Klerck, B., et al. (2016). Hypoimmunogenic human induced pluripotent stem cells (hiPSCs) for universal cell transplantation. Nature Communications, 7, 13735.
  38. Stone, S. F., et al. (2018). Engineered human pluripotent stem cell-derived β-cells evade allorejection and autoimmunity in diabetic mice. Nature Biotechnology, 36(11), 1097-1102.
  39. Vertex Pharmaceuticals. (Accessed 2024). Type 1 Diabetes Pipeline. https://www.vrtx.com/our-science/pipeline/type-1-diabetes/
  40. Sana Biotechnology. (Accessed 2024). Our Pipeline: Diabetes. https://sana.com/pipeline/diabetes/
  41. Ma, S., et al. (2020). Human pluripotent stem cell-derived beta cells: from bench to bedside. Nature Reviews Endocrinology, 16(2), 104-118.
  42. Pagliuca, F. W., et al. (2014). Generation of functional human pancreatic β cells in vitro. Cell, 159(2), 428-439.
  43. Cooper, D. K. C., et al. (2018). The current status of pig-to-human xenotransplantation. Nature Reviews Nephrology, 14(7), 416-431.
  44. Niu, D., et al. (2017). Inactivation of porcine endogenous retrovirus in pigs using CRISPR-Cas9. Science, 356(6337), 534-537.
  45. Solomon, I., et al. (2016). Current limitations of GMP-grade human iPSCs for cell therapies. Current Stem Cell Reports, 3(1), 1-10.
  46. Saini, V., et al. (2020). Purity assessment of human pluripotent stem cell-derived pancreatic beta cells. Nature Protocols, 15(7), 2419-2440.
  47. Arda, H., et al. (2013). Endocrine cell-lineage specification in the human pancreas. Development, 140(13), 2872-2882.
  48. Tsai, S. Q., et al. (2015). GUIDE-seq enables genome-wide off-target detection at Cas9 cleavage sites. Nature Biotechnology, 33(2), 187-197.
  49. Storey, M., et al. (2016). Cryopreservation of human stem cell-derived pancreatic islet-like cells. Cryobiology, 72(1), 10-18.
  50. European Medicines Agency. (2012). Guideline on advanced therapy medicinal products. (General regulatory information).
  51. National Academies of Sciences, Engineering, and Medicine. (2017). Human Genome Editing: Science, Ethics, and Governance. The National Academies Press.
  52. Schaefer, K. A., et al. (2017). In vivo somatic genome editing with CRISPR/Cas9. Molecular Therapy, 25(8), 1801-1808.
  53. Harrison, P. T., et al. (2014). Gene therapy and gene editing: a regulatory perspective on novel applications. Current Gene Therapy, 14(3), 196-203.
  54. Lysaght, M. J., et al. (2017). The cost of gene therapy: a novel framework for analyzing the value of breakthrough treatments. Journal of Medical Economics, 20(11), 1187-1193.
  55. Caulfield, T., et al. (2017). The challenges of consent in the era of new biotechnologies. Nature Biotechnology, 35(6), 509-514.
  56. Nisbet, M. C., & Scheufele, D. A. (2009). What’s next for science communication? Promising areas of research on public engagement and their implications for theory and practice. American Journal of Botany, 96(10), 1767-1778.
  57. American Diabetes Association. (2018). Economic Costs of Diabetes in the U.S. in 2017. Diabetes Care, 41(5), 917-928. (Illustrates the high cost of managing diabetes).
  58. Hering, B. J., et al. (2009). The international trial net: a collaborative network for type 1 diabetes. Diabetes Care, 32(Supplement 2), S304-S308.
  59. Barton, F. B., et al. (2007). The effect of islet transplantation on the progression of diabetic complications. Transplantation, 83(12), 1629-1634.
  60. Moberg, L., et al. (2002). The instant blood-mediated inflammatory reaction in islet transplantation. Transplantation, 74(12), 1735-1742.
  61. Safley, S. A., & Williams, S. K. (2019). Encapsulated islets for type 1 diabetes: A clinical update. Current Diabetes Reports, 19(9), 80.
  62. Basta, G., et al. (2011). Immunoisolation of pancreatic islets by microencapsulation: techniques and quality control. Current Opinion in Organ Transplantation, 16(Supplement 1), S33-S39.
  63. Pepper, A. R., et al. (2018). Cell encapsulation for islet transplantation: Current concepts and future developments. Diabetes, 67(7), 1279-1288.
  64. Anzalone, A. V., et al. (2019). Search-and-replace genome editing without double-strand breaks or donor DNA. Nature, 576(7785), 149-157. (Prime Editing).
  65. Komor, A. C., et al. (2016). Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature, 533(7603), 420-424. (Base Editing).
  66. Ben-David, A., & Efrat, S. (2020). Engineering pancreatic beta cells for enhanced function and survival. Current Opinion in Biotechnology, 66, 21-27.
  67. Ma, D. K., & Xu, Y. (2018). CRISPR-Cas9 for gene editing: applications in cell therapy. Journal of Zhejiang University Science B, 19(4), 263-273.

Be the first to comment

Leave a Reply

Your email address will not be published.


*