Research Report

Enhancing Immunotolerance in Genetically Modified Pigs for Xenotransplantation: Mechanisms and Outcomes  

Tao Zhang
Hangzhou Mono Biotechnology Co., Ltd, Hangzhou, 310000, Zhejiang, China
Author    Correspondence author
International Journal of Molecular Zoology, 2024, Vol. 14, No. 2   doi: 10.5376/ijmz.2024.14.0009
Received: 16 Jan., 2024    Accepted: 25 Feb., 2024    Published: 10 Mar., 2024
© 2024 BioPublisher Publishing Platform
This is an open access article published under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Preferred citation for this article:

Zhang T., 2024, Enhancing immunotolerance in genetically modified pigs for xenotransplantation: mechanisms and outcomes, International Journal of Molecular Zoology, 14(2): 72-83 (doi: 10.5376/ijmz.2024.14.0009)

Abstract

The shortage of human organs for transplantation has driven significant interest in xenotransplantation, particularly using genetically modified pigs. This study explores the mechanisms and outcomes of enhancing immunotolerance in genetically modified pigs for xenotransplantation. Recent advancements in genetic engineering, such as the deletion of xenoantigens and the expression of human complement and coagulation regulatory proteins, have shown promise in reducing immune rejection and prolonging graft survival. Studies have demonstrated that genetically engineered pig hearts and kidneys can survive for extended periods in non-human primates, with some grafts functioning beyond one year. These findings suggest that targeted genetic modifications, combined with specific immunosuppressive regimens, can significantly improve the viability of pig organs for clinical xenotransplantation. However, challenges remain, including the need for further optimization of genetic modifications and immunosuppressive protocols to prevent chronic rejection and ensure long-term graft function.

Keywords
Genetically modified pigs; Xenotransplantation; Immunotolerance; Organ transplantation; Genetic engineering

Xenotransplantation, the transplantation of organs, tissues, or cells from one species to another, has emerged as a promising solution to the critical shortage of human organs available for transplantation. This shortage has led to thousands of patients dying each year while waiting for a suitable donor organ (Sykes and Sachs, 2019). The domestic pig (Sus scrofa domesticus) has been identified as an optimal donor species due to its anatomical and physiological similarities to humans (Klymiuk et al., 2010; Hryhorowicz et al., 2017). However, xenotransplantation faces significant challenges, primarily due to the high immune incompatibility between species, which leads to complex rejection processes (Klymiuk et al., 2010; Hryhorowicz et al., 2017).

 

Recent advancements in genetic engineering, particularly the development of CRISPR-Cas9-based gene editing methodologies, have enabled the creation of genetically modified pigs that are less prone to immune rejection (Sykes and Sachs, 2019). These modifications include the deletion of major xenoantigens, such as alpha-1,3-galactosidase, and the expression of human complement regulatory proteins, coagulation-regulatory proteins, and anti-inflammatory molecules (Klymiuk et al., 2010; Cooper et al., 2019). Such genetic modifications have significantly prolonged the survival of xenografts in preclinical studies, with some genetically engineered pig hearts surviving over a year in non-human primates (Mohiuddin et al., 2014; Yamamoto et al., 2019). These advancements provide hope for the clinical application of xenotransplantation as a viable solution to the organ shortage crisis (Mohiuddin et al., 2014; Sykes and Sachs, 2019).

 

The primary objective of enhancing immunotolerance in genetically modified pigs is to further reduce the immune response and rejection of xenografts, thereby increasing the survival and functionality of transplanted organs. This involves not only the genetic modification of donor pigs but also the development of targeted immunosuppressive therapies to manage the recipient's immune response (Mohiuddin et al., 2014; Yamamoto et al., 2019). By achieving long-term graft survival and function, xenotransplantation could become a routine clinical practice, significantly alleviating the organ shortage and saving countless lives (Mohiuddin et al., 2014; Cooper et al., 2019; Sykes and Sachs, 2019).

 

This study aims to discuss the development of improved xenograft models by leveraging advanced gene editing technology to enhance the immune compatibility of transgenic pigs. The objectives include refining gene editing strategies to improve the adaptability and longevity of pig organs in humans, optimizing immunosuppressive protocols to reduce postoperative risks, and extending the lifespan of transplanted organs. Additionally, the study will address economic and ethical considerations to ensure the responsible development of xenograft technology. The ultimate goal is to establish a robust foundation for translating xenotransplantation from laboratory research to global clinical application, thereby potentially improving the quality of life for transplant patients worldwide.

 

1 Background on Xenotransplantation

1.1 Definition and history of xenotransplantation

Xenotransplantation refers to the transplantation of living cells, tissues, or organs from one species to another. Historically, the concept of xenotransplantation dates back to the early 20th century, with initial attempts involving the transplantation of animal organs into humans. However, these early efforts were largely unsuccessful due to severe immunological rejection and other complications. The advent of modern genetic engineering techniques has reignited interest in xenotransplantation, particularly using pigs as donor animals due to their anatomical and physiological similarities to humans (Hryhorowicz et al., 2017; Lei et al., 2022).

 

1.2 Current challenges in xenotransplantation

Despite significant advancements, xenotransplantation faces several critical challenges. The primary obstacle is immunological rejection, which includes hyperacute rejection (HAR), acute humoral xenograft rejection (AHXR), and chronic rejection. These immune responses are triggered by the recognition of pig-specific antigens by the human immune system (Klymiuk et al., 2010; Carvalho-Oliveira et al., 2021). Additionally, there is a risk of zoonotic infections, particularly from porcine endogenous retroviruses (PERVs), which can potentially be transmitted to human recipients (Klymiuk et al., 2010; Lei et al., 2022). Addressing these challenges is crucial for the successful clinical application of xenotransplantation.

 

1.3 Advances in genetic modification techniques for xenotransplantation

Recent advances in genetic engineering have significantly improved the prospects of xenotransplantation. Techniques such as CRISPR/Cas9 have enabled precise modifications of the pig genome to reduce immunogenicity and enhance compatibility with the human immune system. For instance, the deletion of alpha-1,3-galactosyltransferase (GGTA1) gene, which is responsible for the synthesis of alpha-Gal epitopes, has been shown to mitigate hyperacute rejection (Klymiuk et al., 2010). Additionally, the insertion of human complement and coagulation regulatory genes, such as CD46 and thrombomodulin, has further reduced the risk of immune-mediated rejection and coagulation dysfunctions (Klymiuk et al., 2010; Hryhorowicz et al., 2017; Lei et al., 2022). Moreover, strategies to knock down PERVs have been developed to minimize the risk of zoonotic infections (Klymiuk et al., 2010; Lei et al., 2022). These genetic modifications collectively enhance the immunotolerance of pig organs, making them more viable for xenotransplantation into humans.

 

While xenotransplantation holds great promise for addressing the shortage of human organs, overcoming immunological and infectious challenges remains critical. Advances in genetic modification techniques are paving the way for safer and more effective xenotransplantation practices, bringing us closer to clinical applications (Klymiuk et al., 2010; Hryhorowicz et al., 2017; Carvalho-Oliveira et al., 2021; Lei et al., 2022).

 

2 Immunotolerance: Concepts and Importance

2.1 Definition and mechanisms of immunotolerance

Immunotolerance refers to the immune system's ability to recognize and accept foreign tissues or organs without mounting an aggressive immune response. This is crucial in the context of xenotransplantation, where organs from genetically modified pigs are transplanted into human recipients. The mechanisms of immunotolerance involve both central and peripheral tolerance. Central tolerance occurs in the thymus, where T cells that react strongly to self-antigens are eliminated. Peripheral tolerance involves regulatory T cells (Tregs) and other mechanisms that suppress immune responses to non-self antigens (Shimizu et al., 2012; Carvalho-Oliveira et al., 2021; Lei et al., 2022).

 

2.2 Importance of immunotolerance in the context of xenotransplantation

Immunotolerance is vital for the success of xenotransplantation, as it helps prevent the rejection of transplanted organs. Genetically modified pigs have been engineered to express human complement regulatory proteins and to knock out pig-specific antigens, which helps in reducing the immune response from the human recipient (Fischer et al., 2016; Lei et al., 2022; Xi et al., 2023). Achieving immunotolerance can lead to longer graft survival and better overall outcomes, as demonstrated in studies where genetically modified pig organs showed reduced signs of acute and chronic rejection (Shimizu et al., 2012; Hawthorne et al., 2022). The ability to induce immunotolerance can also minimize the need for long-term immunosuppressive therapy, which has significant side effects (Shimizu et al., 2012; Hawthorne et al., 2022).

 

2.3 Comparison of immunotolerance in human-to-human versus pig-to-human transplants

In human-to-human transplants, immunotolerance is primarily achieved through immunosuppressive drugs and, in some cases, through the induction of donor-specific tolerance via mixed chimerism or Treg therapy. The immune system's response to human antigens is generally less severe compared to xenogeneic antigens from pigs (Shimizu et al., 2012; Lei et al., 2022). In pig-to-human transplants, the immune response is more complex due to the presence of xenogeneic antigens, which can trigger hyperacute rejection, acute vascular rejection, and chronic rejection (Westall et al., 2013; Fischer et al., 2016; Lei et al., 2022). Genetically modified pigs help mitigate these responses by expressing human proteins and knocking out pig-specific antigens, but achieving complete immunotolerance remains a significant challenge (Westall et al., 2013; Fischer et al., 2016; Carvalho-Oliveira et al., 2021; Lei et al., 2022; Xi et al., 2023).

 

Immunotolerance is a critical factor in the success of xenotransplantation. Advances in genetic engineering of pigs have shown promise in reducing immune responses and improving graft survival, but further research is needed to fully understand and achieve long-term immunotolerance in pig-to-human transplants.

 

3 Mechanisms for Enhancing Immunotolerance

3.1 Genetic modifications targeting specific immune pathways

Genetic modifications in pigs have been pivotal in addressing the immunological barriers in xenotransplantation. One of the primary targets is the elimination of xenoantigens, such as α-Gal and Neu5Gc, which are responsible for hyperacute rejection. The knockout of genes like GGTA1 and CMAH has been shown to significantly reduce the binding of human antibodies and subsequent immune responses (Fischer et al., 2016; Butler et al., 2016; Tanihara et al., 2021). Additionally, the insertion of human complement regulatory proteins, such as CD46, CD55, and CD59, has been effective in protecting pig tissues from human complement-mediated lysis (Fischer et al., 2016; Cooper et al., 2019; Lei et al., 2022). These genetic modifications collectively contribute to reducing both humoral and cellular immune responses, thereby enhancing immunotolerance.

 

3.2 Use of CRISPR/Cas9 and other gene editing technologies

The advent of CRISPR/Cas9 technology has revolutionized the field of genetic engineering, allowing for precise and efficient modifications. This technology has been employed to create pigs with multiple gene knockouts, such as GGTA1, CMAH, and B4GALNT2, in a single step, thereby reducing the biosynthesis of xenoantigens (Mohiuddin et al., 2014; Tanihara et al., 2021; Eisenson et al., 2022). CRISPR/Cas9 has also been used to generate pigs with triple knockouts of GGTA1, β2M, and CIITA, which significantly alleviates xenogeneic immune responses by reducing the expression of major histocompatibility complex (MHC) antigens (Fu et al., 2020). The use of CRISPR/Cas9 not only accelerates the generation of genetically modified pigs but also enhances the precision and effectiveness of these modifications, making it a cornerstone technology in xenotransplantation research.

 

3.3 Case studies of successful genetic modifications for immunotolerance

Several case studies have demonstrated the success of genetic modifications in enhancing immunotolerance in xenotransplantation. For instance, pigs with multiple genetic modifications, including the knockout of GGTA1 and the expression of human complement regulatory proteins, have shown complete protection against human complement-mediated lysis and reduced endothelial activation (Fischer et al., 2016). Another study reported the generation of pigs with triple knockouts of GGTA1, β2M, and CIITA, which resulted in significantly prolonged survival of pig skin grafts in immunocompetent mice (Fu et al., 2020) (Figure 1). Additionally, genetically engineered pig hearts expressing human complement and coagulation regulatory genes have achieved long-term survival in baboon models, paving the way for potential clinical applications (Mohiuddin et al., 2014). These case studies underscore the potential of genetic modifications in overcoming immunological barriers and enhancing the success of xenotransplantation.

 

 

Figure 1 The immunogenicity of GGTA1−/−β2M−/−CIITA−/− triple gene knockout (GBC-3KO) pigs in vitro and in vivo (Adopted from Fu et al., 2020)

Image caption: A, Human peripheral blood mononuclear cells (hPBMCs) from healthy volunteers’ blood were labeled with carboxyfluorescein succinimidyl ester (CFSE) and cocultured with irradiated splenocytes (2:1 ratio) from the wild-type (WT) or GBC-3KO fetal pigs for 6 d. Representative data of CFSE density dilution in CD4 and CD8 populations are shown. B, Comparison between WT and GBC-3KO piglet with respect to the CFSE density dilution in CD4 and CD8 populations (n[GBC-3KO] = 6; n[WT] = 3). C, Percentage of CD25 cells in the CD4 and CD8 population of the lymphocytes stimulated by GBC-3KO or WT splenocytes. D, Comparison of the percentage of CD25-positive cells in the CD4 and CD8 population that were stimulated by irradiated splenocytes from the WT or GBC-3KO fetal pigs. E, hPBMCs from healthy volunteers’ blood were cocultured with the irradiated splenocytes (2:1 ratio) from the GBC-3KO or WT fetal pigs for 5 d. Bromodeoxyuridine (BrdU) detection was made by ELISA. The experiment was performed 3×: 3 different GBC-3KO samples and 1 WT sample. F, The absorbance values of 3 experimental results were converted into cell proliferation rate (cell proliferation rate % = experimental well/control well × 100). Each sample has 4 replicates. G, Pig skin grafts (12 × 10 mm) from WT or GBC-3KO pigs were transplanted into C57B6/L mice. Transplant rejection was monitored after day 5. Photographs of the transplanted skin were taken daily. Representative graphs of days 5, 8, 11, and 14 posttransplanted skins are shown. H, Comparison of the survival time of the skin from WT pigs (median survival time [MST], 13.5 d) and from GBC-3KO pigs (MST, 16 d) in C57BL/6 mice. (n[NCG] = 2; n[GBC-TKO] = 15; n[WT] = 10). Plot graphs represent the mean ± SD. Statistical analysis was done using t test or ANOVA with Tukey post hoc test. *P < 0.05, **P < 0.01, ****P < 0.001. HCD8, anti-human CD8 antibody; KO, GBC-KO pig cells with hPBMCs; NCG, the mice that lacking T, B, and NK cells; R, hPBMCs; WT, WT pig cells with hPBMCs (Adopted from Fu et al., 2020)

 

4 Key Genetic Targets for Immunotolerance

4.1 Knockout of xenoantigen genes (e.g., GGTA1, CMAH, β4GalNT2)

The knockout of xenoantigen genes such as GGTA1, CMAH, and β4GalNT2 is a critical strategy to reduce immunogenicity in genetically modified pigs for xenotransplantation. These genes encode enzymes responsible for the synthesis of major carbohydrate antigens, including galactose-α1,3-galactose (αGal), N-glycolylneuraminic acid (Neu5Gc), and Sd(a) antigen, which are recognized by human antibodies and can trigger hyperacute rejection. Studies have demonstrated that the elimination of these antigens significantly reduces human IgG and IgM binding to porcine tissues, thereby minimizing immune responses and improving graft survival (Wang et al., 2018; Zhang et al., 2018; Yoon et al., 2022). For instance, triple knockout (TKO) pigs lacking GGTA1, CMAH, and β4GalNT2 showed minimal human antibody binding and maintained the physical properties of the tissues, making them promising candidates for bioprosthetic heart valves and other xenografts (Zhang et al., 2018; Yoon et al., 2022).

 

4.2 Expression of human regulatory proteins (e.g., CD46, CD55, CD59)

The expression of human regulatory proteins such as CD46, CD55, and CD59 in genetically modified pigs is another essential approach to enhance immunotolerance. These proteins play a crucial role in regulating the complement system, which is part of the innate immune response that can lead to graft rejection. By incorporating these human proteins into pig tissues, researchers aim to inhibit complement activation and reduce immune-mediated damage. Studies have shown that genetically engineered pigs expressing these human complement regulatory proteins exhibit prolonged graft survival and reduced signs of hyperacute rejection in xenotransplantation models (Mohiuddin et al., 2014; Fischer et al., 2016; Burdorf et al., 2021). For example, pig hearts expressing human CD46 and other regulatory proteins have achieved long-term survival in baboon models, indicating the potential for clinical application (Mohiuddin et al., 2014).

 

4.3 Modification of MHC class I and II molecules to reduce immune recognition

Modifying the major histocompatibility complex (MHC) class I and II molecules in pigs is a strategy to reduce immune recognition and subsequent rejection of xenografts. MHC molecules are critical for presenting antigens to T-cells, and their modification can help evade the human immune system. Genetic modifications such as the knockout of swine leukocyte antigen (SLA) class I and II genes, along with the introduction of human leukocyte antigen (HLA) molecules, have been explored to achieve this goal. Research has demonstrated that pigs with reduced or absent SLA expression show decreased T-cell activation and lower levels of human antibody binding, thereby enhancing immunotolerance (Martens et al., 2017; Xu et al., 2022). For instance, pigs with triple gene modifications (GGTA1, B2M, and CIITA) exhibited reduced expression of SLA-I and absence of SLA-II, leading to weaker human immune responses (Xu et al., 2022).

 

4.4 Combined genetic modifications

Combining multiple genetic modifications, including the knockout of xenoantigen genes and the expression of human regulatory proteins, has shown promising results in enhancing immunotolerance. This approach aims to address various aspects of the immune response simultaneously, thereby providing a more comprehensive solution to xenograft rejection. Studies have reported that pigs with combined genetic modifications, such as the knockout of GGTA1 and the expression of human CD46, CD55, and CD59, exhibit significant protection against human complement-mediated lysis and reduced immune activation (Fischer et al., 2016; Yamamoto et al., 2019). These multi-modified pigs represent a significant advancement towards clinical xenotransplantation, demonstrating the potential to overcome the barriers of immune rejection and improve graft survival (Fischer et al., 2016).

 

5 Experimental Outcomes and Clinical Trials

5.1 Summary of preclinical studies and their outcomes

Preclinical studies have demonstrated significant advancements in the field of xenotransplantation using genetically modified pigs. These studies primarily focus on overcoming immune rejection and physiological incompatibilities. For instance, genetically modified pigs with deletions of specific pig genes and insertions of human complement and coagulation-regulatory transgenes have shown promising results in preventing xenograft rejection (Lei et al., 2022). Additionally, the use of porcine neonatal islet cell clusters (NICC) from genetically modified pigs has successfully normalized blood sugar levels in diabetic baboons, with graft survival exceeding 22 months (Hawthorne et al., 2022) (Figure 2). These findings underscore the potential of genetic modifications in enhancing graft survival and function in preclinical models.

 

 

Figure 2 Early morning fasting blood sugar levels (blue) and daily insulin requirements (red) for all recipients (Adopted from Hawthorne et al., 2022)

Image caption: Recipients S1 (A), B1 (B), F1 (C), W1 (D), O1 (E) (Adopted from Hawthorne et al., 2022)

 

5.2 Overview of current and past clinical trials involving genetically modified pigs

Clinical trials involving genetically modified pigs are still in the nascent stages but have shown encouraging results. The first clinical steps have been taken to address the major barriers to xenotransplantation, such as humoral and cellular immune responses and physiological incompatibilities (Lei et al., 2022). In cardiac xenotransplantation, genetically engineered pig hearts have been transplanted into baboons, achieving significant prolongation of graft survival. For example, hearts from genetically engineered piglets expressing human complement regulatory genes and thrombomodulin have survived for over a year in some cases (Mohiuddin et al., 2014). These trials highlight the potential for genetically modified pigs to serve as viable organ donors for human transplantation in the future.

 

5.3 Analysis of immunotolerance and graft survival rates

The analysis of immunotolerance and graft survival rates in xenotransplantation has shown that genetic modifications in donor pigs can significantly enhance graft survival. For instance, the deletion of the αGal gene and the expression of human complement regulators CD55 and CD59 in donor pigs have resulted in long-term diabetes cure in baboons, with graft survival exceeding 22 months (Hawthorne et al., 2022). Similarly, genetically engineered pig hearts with targeted immunosuppression protocols have achieved long-term survival, with some grafts surviving beyond 200 days and even surpassing the one-year mark (Mohiuddin et al., 2014). These outcomes suggest that genetic modifications, combined with appropriate immunosuppressive protocols, can effectively mitigate immune rejection and enhance graft survival in xenotransplantation.

 

6 Challenges and Limitations

6.1 Technical challenges in achieving stable genetic modifications

Achieving stable genetic modifications in pigs for xenotransplantation presents several technical challenges. One primary issue is the complexity of introducing multiple genetic modifications that are necessary to prevent immune rejection and other complications. For instance, pigs need to be genetically engineered to lack certain xenoantigens, such as alpha-1,3-galactosyltransferase, and to express human complement and coagulation regulatory proteins like CD46 and thrombomodulin (Klymiuk et al., 2010; Mohiuddin et al., 2014; Cooper et al., 2019). Ensuring that these modifications are stably integrated and expressed at appropriate levels in the pigs' organs is technically demanding. Additionally, the combination of multiple genetic modifications can lead to unforeseen interactions and complications, making the process even more challenging (Pan et al., 2019; Carvalho-Oliveira et al., 2021).

 

6.2 Potential off-target effects and long-term genetic stability

Another significant challenge is the potential for off-target effects and ensuring long-term genetic stability of the modifications. Techniques such as CRISPR/Cas9, while powerful, can sometimes introduce unintended mutations that may have deleterious effects (Lei et al., 2022; Xi et al., 2023). Moreover, the long-term stability of these genetic modifications is crucial for the success of xenotransplantation. There is a need for extensive in vitro and in vivo testing to ensure that the genetic modifications do not degrade over time or lead to adverse effects in the recipient (Klymiuk et al., 2010; Cooper et al., 2019). The risk of endogenous retroviruses reactivating in genetically modified pigs also poses a long-term genetic stability concern (Pan et al., 2019; Lei et al., 2022).

 

6.3 Ethical and regulatory concerns in the use of genetically modified animals

The use of genetically modified pigs for xenotransplantation raises several ethical and regulatory concerns. Ethical issues revolve around the welfare of the genetically modified animals and the moral implications of using animals for organ harvesting (Mohiuddin et al., 2019; Carvalho-Oliveira et al., 2021). Regulatory bodies require comprehensive evidence to justify the inclusion of each genetic modification, which involves rigorous testing and validation (Cooper et al., 2019; Mohiuddin et al., 2019). Additionally, there are concerns about the potential ecological impact if genetically modified pigs were to interact with wild populations. Regulatory frameworks need to address these issues comprehensively to ensure that the benefits of xenotransplantation do not come at an unacceptable ethical or environmental cost (Cooper et al., 2019; Mohiuddin et al., 2019; Carvalho-Oliveira et al., 2021).

 

7 Ethical and Regulatory Considerations

7.1 Ethical issues related to genetic modification and xenotransplantation

The ethical considerations surrounding the genetic modification of pigs for xenotransplantation are multifaceted. One primary concern is the welfare of the genetically modified animals. The process of genetic modification and subsequent use in xenotransplantation raises questions about the humane treatment of these animals and the potential for suffering. Additionally, there are concerns about the long-term ecological impacts of releasing genetically modified organisms into the environment, even if unintentionally (Pan et al., 2019; Hawthorne et al., 2022; Lei et al., 2022). Another significant ethical issue is the potential for zoonotic diseases. The risk of transferring porcine endogenous retroviruses (PERVs) to human recipients is a major concern, as it could lead to new infectious diseases (Klymiuk et al., 2010; Lei et al., 2022). This risk necessitates rigorous screening and possibly further genetic modifications to mitigate the potential for cross-species disease transmission. Furthermore, the concept of "playing God" by altering the genetic makeup of animals for human benefit is a point of contention. This raises questions about the moral boundaries of scientific intervention and the extent to which humans should manipulate other species for their own needs (Pan et al., 2019; Carvalho-Oliveira et al., 2021).

 

7.2 Regulatory frameworks governing the use of genetically modified pigs

The regulatory landscape for xenotransplantation involving genetically modified pigs is complex and varies by region. In the United States, the Food and Drug Administration (FDA) oversees the regulation of xenotransplantation products, ensuring that they meet stringent safety and efficacy standards before they can be used in clinical settings. This includes comprehensive preclinical testing and ongoing monitoring of recipients for any adverse effects (Klymiuk et al., 2010; Lei et al., 2022). In Europe, the European Medicines Agency (EMA) plays a similar role, with additional oversight from national regulatory bodies. The EMA requires extensive documentation and evidence of safety, including the genetic stability of the modified pigs and the absence of harmful pathogens (Pan et al., 2019). China has also made significant strides in the field of xenotransplantation, with regulatory frameworks that support the development and clinical application of genetically modified pigs. Chinese scientists have been at the forefront of creating multi-gene low-immunogenicity pigs, and their regulatory bodies have established guidelines to ensure the ethical and safe use of these animals in research and clinical trials (Pan et al., 2019).

 

7.3 Public perception and acceptance of xenotransplantation

Public perception and acceptance of xenotransplantation are critical factors that influence the development and implementation of this technology. Public concerns often revolve around the ethical treatment of animals, the potential for disease transmission, and the long-term implications of genetic modifications (Carvalho-Oliveira et al., 2021; Hawthorne et al., 2022; Lei et al., 2022). Educational initiatives and transparent communication from scientists and regulatory bodies are essential to address these concerns and build public trust. Highlighting the potential benefits of xenotransplantation, such as alleviating the shortage of human organs for transplantation and improving patient outcomes, can help garner public support (Klymiuk et al., 2010; Lei et al., 2022). Moreover, cultural and religious beliefs play a significant role in shaping public opinion. Some cultures may have specific objections to the use of pigs or other animals in medical procedures, which must be respectfully considered and addressed through dialogue and engagement with community leaders (Pan et al., 2019; Carvalho-Oliveira et al., 2021). In conclusion, while the potential benefits of xenotransplantation using genetically modified pigs are substantial, addressing the ethical, regulatory, and public perception challenges is crucial for the successful integration of this technology into clinical practice.

 

8 Future Directions and Perspectives

8.1 Emerging trends and future research areas in immunotolerance enhancement

The field of xenotransplantation has seen significant advancements, particularly with the use of genetically modified pigs to overcome immunological barriers. Emerging trends in this area include the development of multi-modified pigs through techniques such as "combineering" and "gene stacking", which allow for the integration of multiple xenoprotective transgenes at a single locus. This approach has shown promise in inhibiting short to mid-term xenograft rejection (Fischer et al., 2016). Additionally, the use of CRISPR/Cas9 technology to disrupt genes responsible for major xenoantigens has been effective in reducing antigenicity and improving pig-to-human compatibility (Li et al., 2021). Future research should focus on refining these genetic modifications and exploring new gene-editing techniques to further enhance immunotolerance.

 

8.2 Potential breakthroughs and technological innovations

Potential breakthroughs in xenotransplantation are likely to come from advancements in both genetic engineering and immunosuppressive therapies. The successful use of genetically modified pig kidneys in brain-dead human recipients, which showed no signs of hyperacute or antibody-mediated rejection, is a significant milestone (Montgomery et al., 2022). Innovations in immunosuppressive regimens, such as the use of anti-CD40 monoclonal antibodies, have also demonstrated improved graft survival in preclinical models (Mohiuddin et al., 2014; Yamamoto et al., 2019). Furthermore, the development of in vitro methods to evaluate human-to-pig xeno-immune responses can accelerate the identification of ideal genetic modifications, thereby expediting the clinical application of xenotransplantation (Li et al., 2021). Continued research in these areas is essential for achieving long-term graft survival and reducing the need for lifelong immunosuppression.

 

8.3 Collaboration opportunities and interdisciplinary approaches

The complexity of xenotransplantation necessitates a multidisciplinary approach, involving collaboration between geneticists, immunologists, transplant surgeons, and bioengineers. Interdisciplinary research can facilitate the development of more effective genetic modifications and immunosuppressive strategies. For instance, combining expertise in gene editing with advancements in immunology has already led to the creation of genetically engineered pigs that express human complement regulatory genes, significantly improving graft survival (Yang and Sykes, 2007; Fischer et al., 2016). Collaborative efforts should also focus on addressing the remaining physiological incompatibilities and zoonotic risks associated with xenotransplantation (Lu et al., 2020; Lei et al., 2022). By fostering partnerships across various scientific disciplines, the field can move closer to the goal of clinical application and ultimately solve the organ shortage crisis.

 

In summary, the future of xenotransplantation lies in the continued refinement of genetic modifications, the development of innovative immunosuppressive therapies, and the fostering of interdisciplinary collaborations. These efforts will pave the way for successful clinical trials and the eventual widespread use of xenotransplantation to address the critical shortage of human organs.

 

9 Concluding Remarks

The research on enhancing immunotolerance in genetically modified pigs for xenotransplantation has made significant strides. Key genetic modifications, such as the deletion of carbohydrate xenoantigens and the expression of human complement and coagulation-regulatory proteins, have been shown to improve graft survival and function. For instance, pigs with nine genetic modifications, including the deletion of all three known carbohydrate xenoantigens and the expression of human CD46, CD55, thrombomodulin, endothelial cell protein C receptor, human hemeoxygenase-1, and human CD47, have demonstrated promising results in preclinical studies. These modifications help mitigate hyperacute rejection, acute humoral xenograft rejection, and immune cell-mediated rejection, which are major barriers to successful xenotransplantation.

 

The findings suggest that genetically modified pigs could potentially serve as viable organ donors for humans, addressing the critical shortage of human organs for transplantation. Future research should focus on optimizing the combination of genetic modifications to further enhance graft survival and function. Additionally, clinical trials are necessary to validate the efficacy and safety of these genetically modified pig organs in human recipients. The development of more targeted immunosuppressive therapies, as demonstrated by the prolonged survival of pig heart xenografts in baboons, also holds promise for improving clinical outcomes.

 

Continued research is essential to refine genetic modifications and immunosuppressive protocols to ensure the long-term success of xenotransplantation. Researchers and clinicians must work together to address the remaining physiological and immunological challenges. Moreover, ethical considerations, including the welfare of genetically modified pigs and the potential risks to human recipients, must be carefully evaluated. Regulatory frameworks should be established to oversee the ethical and safe implementation of xenotransplantation in clinical practice. In conclusion, while significant progress has been made, ongoing research and ethical vigilance are crucial to realizing the full potential of xenotransplantation in addressing the organ shortage crisis.

 

Acknowledgements

The author extends sincere thanks to two anonymous peer reviewers for their invaluable feedback on the manuscript of this paper, whose critical evaluations and constructive suggestions have greatly contributed to the improvement of this manuscript.

 

Conflict of Interest Disclosure

The author affirms that this research was conducted without any commercial or financial relationships that could be construed as a potential conflict of interest.

 

References 

Burdorf L., Laird C., Harris D., Connolly M., Habibabady Z., Redding E., O'Neill N., Cimeno A., Parsell D., Phelps C., Ayares D., Azimzadeh A., and Pierson R., 2021, Pig‐to‐baboon lung xenotransplantation: extended survival with targeted genetic modifications and pharmacologic treatments, American Journal of Transplantation, 22(1): 28-45.

https://doi.org/10.1111/ajt.16809
PMid:34424601 PMCid:PMC10292947

 

Butler J., Wang Z., Martens G., Ladowski J., Li P., Tector M., and Tector A., 2016, Modified glycan models of pig-to-human xenotransplantation do not enhance the human-anti-pig T cell response, Transplant immunology, 35: 47-51.

https://doi.org/10.1016/j.trim.2016.02.001
PMid:26873419

 

Carvalho-Oliveira M., Valdivia E., Blasczyk R., and Figueiredo C., 2021, Immunogenetics of xenotransplantation, International Journal of Immunogenetics, 48(2): 120-134.

https://doi.org/10.1111/iji.12526
PMid:33410582

 

Cooper D., Hara H., Iwase H., Yamamoto T., Li Q., Ezzelarab M., Federzoni E., Dandro A., and Ayares D., 2019, Justification of specific genetic modifications in pigs for clinical organ xenotransplantation, Xenotransplantation, 26(4): e12516.

https://doi.org/10.1111/xen.12516
PMid:30989742 PMCid:PMC10154075

 

Eisenson D., Hisadome Y., and Yamada K., 2022, Progress in xenotransplantation: immunologic barriers, advances in gene editing, and successful tolerance induction strategies in pig-to-primate transplantation, Frontiers in Immunology, 13: 899657.

https://doi.org/10.3389/fimmu.2022.899657
PMid:35663933 PMCid:PMC9157571

 

Fischer K., Kraner-Scheiber S., Petersen B., Rieblinger B., Buermann A., Flisikowska T., Flisikowski K., Christan S., Edlinger M., Baars W., Kurome M., Zakhartchenko V., Kessler B., Plotzki E., Szczerbal I., Świtoński M., Denner J., Wolf E., Schwinzer R., Niemann H., Kind A., and Schnieke A., 2016, Efficient production of multi-modified pigs for xenotransplantation by ‘combineering’, gene stacking and gene editing, Scientific Reports, 6(1): 29081.

https://doi.org/10.1038/srep29081
PMid:27353424 PMCid:PMC4926246

 

Fu R., Fang M., Xu K., Ren J., Zou J., Su L., Chen X., An P., Yu D., Ka M., Hai T., Li Z., Li W., Yang Y., Zhou Q., and Hu Z., 2020, Generation of GGTA1-/-β2M-/-CIITA-/-pigs using CRISPR/Cas9 technology to alleviate xenogeneic immune reactions, Transplantation, 104(8): 1566-1573.

https://doi.org/10.1097/TP.0000000000003205
PMid:32732833

 

Hawthorne W., Salvaris E., Chew Y., Burns H., Hawkes J., Barlow H., Hu M., Lew A., Nottle M., O’Connell P., and Cowan P., 2022, Xenotransplantation of genetically modified neonatal pig islets cures diabetes in baboons, Frontiers in Immunology, 13: 898948.

https://doi.org/10.3389/fimmu.2022.898948
PMid:35784286 PMCid:PMC9243461

 

Hryhorowicz M., Zeyland J., Słomski R., and Lipiński D., 2017, Genetically modified pigs as organ donors for xenotransplantation, Molecular Biotechnology, 59: 435-444.

https://doi.org/10.1007/s12033-017-0024-9
PMid:28698981 PMCid:PMC5617878

 

Klymiuk N., Aigner B., Brem G., and Wolf E., 2010, Genetic modification of pigs as organ donors for xenotransplantation, Molecular Reproduction and Development, 77(3): 209-221.

https://doi.org/10.1002/mrd.21127
PMid:19998476

 

Lei T., Chen L., Wang K., Du S., Gonelle-Gispert C., Wang Y., and Buhler L., 2022, Genetic engineering of pigs for xenotransplantation to overcome immune rejection and physiological incompatibilities: the first clinical steps, Frontiers in Immunology, 13: 1031185.

https://doi.org/10.3389/fimmu.2022.1031185
PMid:36561750 PMCid:PMC9766364

 

Li P., Walsh J., Lopez K., Isidan A., Zhang W., Chen A., Goggins W., Higgins N., Liu J., Brutkiewicz R., Smith L., Hara H., Cooper D., and Ekser B., 2021, Genetic engineering of porcine endothelial cell lines for evaluation of human-to-pig xenoreactive immune responses, Scientific Reports, 11(1): 13131.

https://doi.org/10.1038/s41598-021-92543-y
PMid:34162938 PMCid:PMC8222275

 

Lu T., Yang B., Wang R., and Qin C., 2020, Xenotransplantation: current status in preclinical research, Frontiers in Immunology, 10: 3060.

https://doi.org/10.3389/fimmu.2019.03060
PMid:32038617 PMCid:PMC6989439

 

Martens G., Reyes L., Li P., Butler J., Ladowski J., Estrada J., Sidner R., Eckhoff D., Tector M., and Tector A., 2017, Humoral reactivity of renal transplant-waitlisted patients to cells From GGTA1/CMAH/B4GalNT2, and SLA class I knockout pigs, Transplantation, 101(4): e86-e92.

https://doi.org/10.1097/TP.0000000000001646
PMid:28114170 PMCid:PMC7228580

 

Mohiuddin M., Singh A., Corcoran P., Hoyt R., Thomas M., Ayares D., and Horvath K., 2014, Genetically engineered pigs and target-specific immunomodulation provide significant graft survival and hope for clinical cardiac xenotransplantation, The Journal of Thoracic and Cardiovascular Surgery, 148(3): 1106-13; discussion 1113-4.

https://doi.org/10.1016/j.jtcvs.2014.06.002
PMid:24998698 PMCid:PMC4135017

 

Mohiuddin M., DiChiacchio L., Singh A., and Griffith B., 2019, Xenotransplantation: a step closer to clinical reality? Transplantation, 103(3): 453-454.

https://doi.org/10.1097/TP.0000000000002608
PMid:30801425

 

Montgomery R., Stern J., Lonze B., Tatapudi V., Mangiola M., Wu M., Weldon E., Lawson N., Deterville C., Dieter R., Sullivan B., Boulton G., Parent B., Piper G., Sommer P., Cawthon S., Duggan E., Ayares D., Dandro A., Fazio-Kroll A., Kokkinaki M., Burdorf L., Lorber M., Boeke J., Pass H., Keating B., Griesemer A., Ali N., Mehta S., and Stewart Z., 2022, Results of two cases of pig-to-human kidney xenotransplantation, The New England Journal of Medicine, 386(20): 1889-1898.

https://doi.org/10.1056/NEJMoa2120238
PMid:35584156

 

Pan D., Liu T., Lei T., Zhu H., Wang Y., and Deng S., 2019, Progress in multiple genetically modified minipigs for xenotransplantation in China, Xenotransplantation, 26(1): e12492.

https://doi.org/10.1111/xen.12492
PMid:30775816

 

Shimizu A., Yamada K., Robson S., Sachs D., and Colvin R., 2012, Pathologic characteristics of transplanted kidney xenografts, Journal of the American Society of Nephrology, 23(2): 225-235.

https://doi.org/10.1681/ASN.2011040429
PMid:22114174 PMCid:PMC3269172

 

Sykes M., and Sachs D., 2019, Transplanting organs from pigs to humans, Science Immunology, 4(41): eaau6298.

https://doi.org/10.1126/sciimmunol.aau6298
PMid:31676497 PMCid:PMC7293579

 

Tanihara F., Hirata M., Nguyen N., Sawamoto O., Kikuchi T., and Otoi T., 2021, One-step generation of multiple gene-edited pigs by electroporation of the CRISPR/Cas9 system into zygotes to reduce xenoantigen biosynthesis, International Journal of Molecular Sciences, 22(5): 2249.

https://doi.org/10.3390/ijms22052249
PMid:33668187 PMCid:PMC7956194

 

Wang R., Ruan M., Zhang R., Chen L., Li X., Fang B., Li C., Ren X., Liu J., Xiong Q., Zhang L., Jin Y., Li L., Li R., Wang Y., Yang H., and Dai Y., 2018, Antigenicity of tissues and organs from GGTA1/CMAH/β4GalNT2 triple gene knockout pigs, The Journal of Biomedical Research, 33(4): 235-243..

https://doi.org/10.7555/JBR.32.20180018
PMid:30007952 PMCid:PMC6813527

 

Westall G., Levvey B., Salvaris E., Gooi J., Marasco S., Rosenfeldt F., Egan C., Ccp R., Mennen M., Russell P., Robson S., Nottle M., Dwyer K., Snell G., and Cowan P., 2013, Sustained function of genetically modified porcine lungs in an ex vivo model of pulmonary xenotransplantation, The Journal of Heart and Lung Transplantation, 32(11): 1123-1130.

https://doi.org/10.1016/j.healun.2013.07.001
PMid:23932853

 

Xi J., Zheng W., Chen M., Zou Q., Tang C., and Zhou X., 2023, Genetically engineered pigs for xenotransplantation: Hopes and challenges, Frontiers in Cell and Developmental Biology, 10: 1093534.

https://doi.org/10.3389/fcell.2022.1093534
PMid:36712969 PMCid:PMC9878146

 

Xu K., Yu H., Chen S., Zhang Y., Guo J., Yang C., Jiao D., Nguyen T., Zhao H., Wang J., Wei T., Li H., Jia B., Jamal M., Zhao H., Huang X., and Wei H., 2022, Production of triple-gene (GGTA1, B2M and CIITA)-modified donor pigs for xenotransplantation, Frontiers in Veterinary Science, 9: 848833.

https://doi.org/10.3389/fvets.2022.848833
PMid:35573408 PMCid:PMC9097228

 

Yamamoto T., Hara H., Foote J., Wang L., Li Q., Klein E., Schuurman H., Zhou H., Li J., Tector A., Zhang Z., Ezzelarab M., Lovingood R., Ayares D., Eckhoff D., Cooper D., and Iwase H., 2019, Life-supporting kidney xenotransplantation from genetically-engineered pigs in baboons: a comparison of two immunosuppressive regimens, Transplantation, 103(10): 2090-2104.

https://doi.org/10.1097/TP.0000000000002796
PMid:31283686

 

Yang Y., and Sykes M., 2007, Xenotransplantation: current status and a perspective on the future, Nature Reviews Immunology, 7(7): 519-531.

https://doi.org/10.1038/nri2099
PMid:17571072

 

Yoon S., Lee S., Park C., Choi H., Yoo M., Lee S., Hyun C., Kim N., Kang T., Son E., Ghosh M., Son Y., and Hur C., 2022, An efficacious transgenic strategy for triple knockout of xeno-reactive antigen genes GGTA1, CMAH, and B4GALNT2 from Jeju native pigs, Vaccines, 10(9): 1503.

https://doi.org/10.3390/vaccines10091503
PMid:36146581 PMCid:PMC9505423

 

Zhang R., Wang Y., Chen L., Wang R., Li C., Li X., Fang B., Ren X., Ruan M., Liu J., Xiong Q., Zhang L., Jin Y., Zhang M., Liu X., Li L., Chen Q., Pan D., Li R., Cooper D., Yang H., and Dai Y., 2018, Reducing immunoreactivity of porcine bioprosthetic heart valves by genetically-deleting three major glycan antigens, GGTA1/β4GalNT2/CMAH, Acta Biomaterialia, 72: 196-205.

https://doi.org/10.1016/j.actbio.2018.03.055
PMid:29631050

 

International Journal of Molecular Zoology
• Volume 14
View Options
. PDF(1175KB)
. FPDF(win)
. FPDF(mac)
. HTML
. Online fPDF
Associated material
. Readers' comments
Other articles by authors
. Tao Zhang
Related articles
. Genetically modified pigs
. Xenotransplantation
. Immunotolerance
. Organ transplantation
. Genetic engineering
Tools
. Email to a friend
. Post a comment