Feature Review

Genetic Determinants of Long-Term Graft Survival in Pig-to-Human Xenotransplantation  

Jessi J. White
Tropical Animal Medicine Center, Hainan Institute of Tropical Agricultural Resources, Sanya, 572024, Hainan, China
Author    Correspondence author
International Journal of Molecular Medical Science, 2024, Vol. 14, No. 3   
Received: 15 May, 2024    Accepted: 18 Jun., 2024    Published: 28 Jun., 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.
Abstract

Xenotransplantation technology, with its potential contribution to addressing the human organ shortage, is gradually becoming a hotspot in medical research. This study focuses on analyzing the genetic factors affecting the long-term survival of xenografts, with a particular emphasis on reducing immune rejection responses and enhancing graft-host compatibility through genetic modification. The application of CRISPR/Cas9 technology allows for the precise knockout of porcine antigens such as GGTA1, CMAH, and β4GalNT2, as well as the introduction of human complement regulatory proteins like CD46, CD55, and thrombomodulin, which have shown significant effectiveness in mitigating rejection reactions. Additionally, this study extensively discusses the technical, ethical, and regulatory challenges faced by xenotransplantation technology and emphasizes the necessity of interdisciplinary research and collaboration. This research not only provides a scientific basis for the clinical application of xenogeneic organ transplantation but also has profound implications for promoting interdisciplinary cooperation, fostering in-depth discussions on medical ethics, and enhancing public understanding of biotechnological applications.

Keywords
Xenotransplantation; Genetic modifications; Graft survival; Immunosuppression; Pig-to-human transplantation; Gene editing

1 Introduction

Xenotransplantation, the process of transplanting organs, tissues, or cells from one species to another, is an innovative approach to addressing the acute shortage of human organs available for transplantation. The most prominent focus in xenotransplantation research has been on pig-to-human transplants due to the anatomical and physiological similarities between pigs and humans. These similarities make pigs an ideal source for organs such as kidneys, hearts, and livers, which are in high demand for transplantation.

 

The significance of xenotransplantation lies in its potential to drastically reduce the waiting time for organ transplants. Currently, many patients die while waiting for suitable human donors. Xenotransplantation offers a virtually unlimited supply of donor organs, which could meet the growing demand and save countless lives. Additionally, it provides a consistent and controlled source of organs, which can be genetically modified to reduce the risk of rejection and improve compatibility with human recipients (Cooper et al., 2020).

 

Pigs are particularly suitable for xenotransplantation for several reasons. Firstly, pigs have large litters and short gestation periods, making them readily available and easy to breed. Secondly, the size and function of pig organs are similar to human organs, which facilitates the transplantation process. Furthermore, advancements in genetic engineering have enabled scientists to modify pig genomes to remove or alter genes that trigger immune responses in humans. This genetic modification helps in reducing the risk of hyperacute rejection, a major barrier in xenotransplantation (Tatapudi and Griesemer, 2022).

 

The potential benefits of xenotransplantation extend beyond individual patient outcomes. It also has significant implications for public health and the healthcare system. By providing a steady supply of organs, xenotransplantation can alleviate the burden on the healthcare system associated with long-term dialysis for kidney failure patients or the extensive medical management required for patients with end-stage organ failure. Additionally, it can reduce the economic costs associated with prolonged hospital stays and repeated medical interventions for patients awaiting transplants.

 

Despite its potential, pig-to-human xenotransplantation faces numerous challenges that must be addressed to achieve successful and long-term graft survival. These challenges include hyperacute rejection, where the human immune system rapidly attacks the transplanted pig organ, as well as chronic rejection, which can occur over a longer period. Additionally, there is a risk of zoonotic infections, where diseases can be transmitted from pigs to humans. The primary goal of pig-to-human organ transplants is to overcome these immunological barriers and ensure that the transplanted organs can function effectively and safely in the human body over the long term (Burdorf et al., 2018; Reichart et al., 2020).

 

The primary objective of this study is to elucidate the genetic determinants that contribute to the long-term survival of xenografts from pigs to humans. Understanding these genetic factors is crucial for optimizing the genetic modification of donor pigs to enhance graft survival and function. By thoroughly reviewing the current literature on the genetic engineering of pigs for xenotransplantation, the most effective genetic modifications and their impacts on overcoming immune rejection and physiological incompatibilities are identified. This study will provide a foundation for future research and clinical trials, ultimately advancing the field of xenotransplantation and offering new hope to patients in need of life-saving organ transplants.

 

2 Background on Xenotransplantation

2.1 Definition and historical context

Xenotransplantation, defined as the transplantation of living cells, tissues, or organs from one species to another, has a long and complex history. The term "xeno" is derived from the Greek word for "foreign," highlighting the fundamental challenge of overcoming species-specific biological differences. The earliest recorded attempts at xenotransplantation date back to the early 20th century when researchers explored the potential of using animal organs to address the shortage of human donors.

 

One of the first significant attempts was by French surgeon Alexis Carrel, who, in the 1900s, experimented with transplanting animal organs into humans. However, these early experiments faced numerous obstacles, primarily due to the body's immune response, which led to immediate and catastrophic rejection of the transplanted organs. The lack of understanding of immunology and the absence of effective immunosuppressive therapies at the time resulted in these early attempts being largely unsuccessful (Gulyaev et al., 2019).

 

The mid-20th century saw renewed interest in xenotransplantation with advancements in surgical techniques and a better understanding of transplantation immunology. Notably, in the 1960s, Dr. Keith Reemtsma at Tulane University transplanted chimpanzee kidneys into human patients. Although one patient survived for nine months, the overall outcomes were poor due to immune rejection and infections. These experiments underscored the need for more sophisticated immunosuppressive strategies and a better understanding of cross-species immunological barriers (Hess and Kaczorowski, 2023).

 

The 1980s brought further attention to xenotransplantation with the case of Baby Fae, an infant born with a fatal heart defect who received a baboon heart transplant. Despite initial success, the infant's body rejected the heart after 21 days, leading to her death. This case highlighted both the potential and the significant challenges of xenotransplantation, particularly the need for better immunosuppressive treatments and the ethical considerations involved in such procedures (Loike and Kadish, 2018).

 

In recent decades, the focus has shifted towards the use of pigs as the preferred source of xenotransplant organs due to their anatomical and physiological similarities to humans. Advances in genetic engineering have enabled the development of transgenic pigs that express human proteins to mitigate immune rejection. For instance, the knockout of the alpha-1,3-galactosyltransferase (Gal) gene in pigs, which eliminates a major xenoantigen responsible for hyperacute rejection, has been a pivotal breakthrough (Cyprian, 2020).

 

2.2 Current advancements and breakthroughs in xenotransplantation

Recent years have witnessed significant advancements in xenotransplantation, primarily driven by breakthroughs in genetic engineering and immunosuppression techniques. One of the most pivotal developments has been the creation of genetically modified pigs that are specifically engineered to be more compatible with the human immune system. These pigs have undergone modifications to eliminate or alter the expression of certain pig antigens that are typically recognized and attacked by the human immune system. Notably, the knockout of the alpha-1,3-galactosyltransferase (Gal) gene in pigs has been a major milestone, as this gene is responsible for producing a sugar molecule that is a primary target for human antibodies. Additionally, the expression of human complement regulatory proteins such as CD46, CD55, and thrombomodulin in these pigs helps to further protect the transplanted organs from immune attack by inhibiting the complement cascade, a part of the immune system involved in inflammation and cell lysis (Singh et al., 2018; Coe et al., 2020).

 

In addition to genetic modifications, advancements in immunosuppressive therapies have played a crucial role in the success of xenotransplantation. Traditional immunosuppressive drugs used in allotransplantation have been supplemented with novel regimens designed specifically to address the unique challenges of xenotransplantation. Co-stimulation blockade therapies, which inhibit the interaction between co-stimulatory molecules on immune cells, have shown great promise in reducing immune responses against xenografts. For instance, antibodies targeting the CD40-CD154 pathway have been effective in prolonging graft survival by preventing the activation of T cells, which are essential for initiating immune responses. These advancements have led to significant improvements in the survival rates of xenotransplanted organs in preclinical studies. For example, genetically modified pig kidneys transplanted into non-human primates have shown survival times extending into months, marking a substantial improvement over earlier attempts (Reichart et al., 2020; Hawthorne et al., 2022). These breakthroughs highlight the potential of xenotransplantation to become a viable clinical option, pending further research and refinement of these techniques.

 

2.3 Comparison of xenotransplantation with allotransplantation

Xenotransplantation offers several potential advantages over traditional allotransplantation, which involves the transfer of organs or tissues between individuals of the same species. The most significant advantage is the potential to alleviate the severe shortage of human donor organs. Genetically modified pigs can be bred in large numbers, providing a consistent and readily available source of organs. This capability could dramatically reduce waiting times for patients in need of transplants and decrease mortality rates associated with long waiting periods. Additionally, the controlled breeding of pigs allows for specific genetic modifications to enhance compatibility and reduce rejection rates, which is not possible with human donors (Cooper et al., 2020; Reichart et al., 2020).

 

However, xenotransplantation faces unique challenges that are less prevalent or absent in allotransplantation. One of the primary challenges is the risk of zoonotic infections, where diseases could potentially be transmitted from pigs to humans. While significant progress has been made in producing pathogen-free pigs, this remains a critical concern. Furthermore, the human immune system often mounts a stronger rejection response against pig tissues compared to human tissues, necessitating more complex and robust immunosuppressive therapies. Despite the success of these therapies in preclinical trials, their long-term efficacy and safety in humans remain uncertain. In contrast, allotransplantation has a well-established history with protocols that have been refined over decades to manage immune rejection and ensure long-term graft survival. Thus, while xenotransplantation holds immense promise for addressing organ shortages, it must overcome substantial scientific and medical hurdles to match the reliability and success of allotransplantation (Burdorf et al., 2018; Firl and Markmann, 2022).

 

3 Immunological Barriers in Xenotransplantation

3.1 Host immune response to xenografts

The host immune response to xenografts is a significant barrier in xenotransplantation. The immune system recognizes the xenograft as foreign, leading to a robust immune response aimed at rejecting the transplanted organ. This response involves both humoral and cellular components of the immune system. Humoral responses are primarily mediated by antibodies that target specific antigens on the xenograft, while cellular responses involve T cells that attack the graft tissue. Studies have shown that genetically modified pigs, which lack certain antigens or express human regulatory proteins, can mitigate these immune responses to some extent (Firl and Markmann, 2022; Lei et al., 2022; Wu et al., 2023). This response involves both the innate and adaptive immune systems, leading to various types of graft rejections. Key components of this immune response include natural antibodies, particularly those targeting the alpha-Gal antigen, and complement activation, which contribute to the rapid rejection of the xenograft (Singh et al., 2018).

 

3.2 Hyperacute rejection, acute vascular rejection, and chronic rejection

Xenotransplantation faces several types of graft rejection, each posing significant challenges to the long-term success of pig-to-human transplants (Fiugre 1). Understanding the mechanisms and manifestations of these rejections is crucial for developing effective strategies to prevent and manage them.

 

1) Hyperacute rejection:

Hyperacute rejection (HAR) is the most immediate and severe form of xenograft rejection, occurring within minutes to hours after transplantation. It is primarily mediated by pre-existing natural antibodies in the recipient's blood, which recognize specific antigens on the surface of the xenograft, such as the alpha-Gal epitope. These antibodies activate the complement system, leading to a cascade of events that result in rapid and extensive damage to the graft’s endothelial cells. This causes the blood vessels within the graft to become occluded by clots, resulting in ischemia and necrosis of the transplanted tissue. The rapid onset and severity of HAR make it a major obstacle in xenotransplantation (Singh et al., 2018; Tatapudi and Griesemer, 2022).

 

2) Acute vascular rejection:

Acute vascular rejection (AVR), also known as delayed xenograft rejection, typically occurs days to weeks after transplantation. AVR is characterized by an immune response that targets the blood vessels of the xenograft, leading to endothelial cell activation and inflammation. This results in vascular injury, thrombosis, and eventual graft failure. Key players in AVR include activated macrophages, natural killer cells, and T cells, which infiltrate the graft and contribute to the inflammatory milieu. The presence of these immune cells and the associated inflammatory response lead to swelling, hemorrhage, and compromised function of the xenograft (Coe et al., 2020; Reichart et al., 2020).

 

3) Chronic rejection:

Chronic rejection is a long-term process that develops over months to years post-transplantation. Unlike hyperacute and acute vascular rejection, chronic rejection involves both immune and non-immune mechanisms. It is characterized by progressive fibrosis, vascular occlusion, and a gradual decline in graft function. Chronic rejection is driven by a persistent, low-grade immune response, involving both cellular and humoral components. T cells and antibodies continuously attack the graft, leading to sustained inflammation and tissue remodeling. Additionally, non-immune factors such as ischemia, reperfusion injury, and drug toxicity contribute to the pathogenesis of chronic rejection. The slow and insidious nature of chronic rejection makes it difficult to diagnose and treat, often resulting in eventual graft failure (Burdorf et al., 2018; Firl and Markmann, 2022).

 

3.3 Role of genetic modifications in overcoming immunological barriers

Genetic modifications in donor pigs have been instrumental in mitigating the immunological barriers to xenotransplantation. Key strategies include:

 

Knockout of Xenoantigens: The deletion of genes encoding major xenoantigens, such as alpha-Gal, has been crucial in reducing hyperacute rejection. Pigs genetically modified to lack the alpha-Gal antigen have shown significantly improved graft survival (Reichart et al., 2020).

 

Expression of Human Complement Regulatory Proteins: Transgenic expression of human proteins such as CD46, CD55, and thrombomodulin in pigs helps protect the graft from complement-mediated damage. These proteins regulate the complement cascade and prevent excessive immune activation (Singh et al., 2018).

 

Coagulation and Inflammation Control: Genetic modifications to include human thromboregulatory proteins and anti-inflammatory genes have also been shown to reduce the incidence of acute vascular rejection and improve overall graft survival. These modifications help in maintaining the normal physiological function of the graft and reducing inflammatory responses (Burdorf et al., 2018).

 

Immune Modulation through Costimulation Blockade: Genetically modified pigs can be used in conjunction with advanced immunosuppressive therapies, such as costimulation blockade. This involves using antibodies that block critical pathways in T-cell activation, such as the CD40-CD154 interaction. Studies have shown that the combination of genetically modified pigs and costimulation blockade can significantly prolong graft survival and reduce the risk of acute rejection (Firl and Markmann, 2022).

 

Development of Multi-Transgenic Pigs: The use of multi-transgenic pigs, which incorporate multiple genetic modifications to address various aspects of the immune response, has shown promising results. These pigs may express combinations of human regulatory proteins, lack multiple xenoantigens, and include additional modifications to enhance graft survival. Such comprehensive genetic engineering efforts are essential for overcoming the complex immunological barriers in xenotransplantation (Tatapudi and Griesemer, 2022).

 

In summary, overcoming the immunological barriers to xenotransplantation requires a multifaceted approach, incorporating advanced genetic modifications and tailored immunosuppressive therapies. These strategies hold the promise of significantly enhancing the success rates of pig-to-human xenotransplantations.

 

4 Key Genetic Determinants for Graft Survival

4.1 Genes involved in immune evasion and tolerance

The success of xenotransplantation largely hinges on the ability to evade the host's immune system and promote tolerance. Several genes have been identified that play critical roles in immune evasion and tolerance, including those that modulate the expression of surface antigens and immune regulatory proteins. For instance, the expression of human leukocyte antigen-G (HLA-G) and HLA-E in genetically modified pigs has been shown to suppress natural killer (NK) cell activity and promote immune tolerance (Obando et al., 2021; Lopez et al., 2022). Moreover, the introduction of porcine vascularized thymic grafts has shown promise in inducing tolerance by re-educating the recipient’s immune system to accept the xenograft (Yamada et al., 2020).

 

One promising approach involves the use of immune-privileged cells, such as neonatal pig Sertoli cells (NPSC), which have been shown to create an immune modulatory environment. These cells can prolong the survival of co-transplanted cells by recruiting regulatory T cells (Tregs) and producing immunoregulatory factors like TGF-β and IL-10, which help in reducing inflammation and apoptosis (Kaur et al., 2020). Additionally, the deletion of genes such as β2-microglobulin (β2M) and CIITA in pigs has been shown to reduce the activation and proliferation of human T cells, thereby alleviating xenogeneic immune responses and prolonging graft survival (Fu et al., 2020).

 

4.2 Genetic modifications to reduce antigenicity

One of the primary barriers to successful xenotransplantation is the presence of xenoantigens on pig tissues, which can trigger hyperacute rejection. The most significant xenoantigens include galactose-alpha-1,3-galactose (α-Gal), N-glycolylneuraminic acid (Neu5Gc), and the Sd(a) antigen. Genetic modifications to knock out these antigens have shown promising results. For instance, pigs with triple gene knockouts (GGTA1, CMAH, and β4GalNT2) exhibit significantly reduced antigenicity, as evidenced by decreased human IgG and IgM binding to their tissues (Wang et al., 2018; Yoon et al., 2022). This reduction in antigenicity is crucial for minimizing hyperacute rejection and improving graft survival.

 

4.3 Genes enhancing organ resistance to rejection and injury

In addition to reducing antigenicity, enhancing the graft’s intrinsic resistance to immune-mediated damage is essential. Genes encoding human complement regulatory proteins, such as CD46, CD55, and CD59, have been introduced into pigs to protect against complement-mediated lysis. These modifications help in preventing acute vascular rejection and improving long-term graft survival (Singh et al., 2018; Lei et al., 2022). Moreover, genetic modifications to enhance the expression of anti-inflammatory and anti-apoptotic genes in pigs have shown to reduce tissue injury and improve graft durability. For example, pigs expressing human thrombomodulin have demonstrated significantly prolonged graft survival by reducing coagulation-related complications (Porrett et al., 2023).

 

5 CRISPR/Cas9 and Genetic Engineering in Xenotransplantation

5.1 Overview of CRISPR/Cas9 technology and its applications

CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats associated with Cas9) is a revolutionary gene-editing technology that allows for precise modifications of DNA within organisms. The system consists of the Cas9 nuclease, which cuts DNA at specific locations guided by a synthetic RNA molecule that matches the target DNA sequence. This technology has transformed genetic research and biotechnology, enabling targeted gene knockouts, insertions, and modifications with high efficiency and accuracy. CRISPR/Cas9 has been widely adopted for various applications, including biomedical research, agricultural improvements, and the development of genetically modified organisms. In the context of xenotransplantation, CRISPR/Cas9 is employed to modify the genomes of donor pigs to make their organs more compatible with human recipients (Ryczek et al., 2021).

 

In the context of xenotransplantation, CRISPR/Cas9 has been employed to modify the genomes of donor pigs to reduce the risk of immune rejection and improve graft survival. By knocking out specific genes that are responsible for hyperacute rejection, such as the alpha-1,3-galactosyltransferase (GGTA1) gene, researchers have been able to create genetically modified pigs that are more compatible with human immune systems (Firl and Markmann, 2022; Montgomery et al., 2022).

 

5.2 Case studies of CRISPR/Cas9-mediated genetic modifications in pigs

Several case studies highlight the successful application of CRISPR/Cas9 in creating genetically modified pigs for xenotransplantation:

 

Tanihara et al. (2021) utilized CRISPR/Cas9 to simultaneously knock out three key genes involved in xenoantigen biosynthesis—GGTA1, CMAH, and B4GALNT2. This approach aimed to reduce the antigenicity of pig organs, thereby minimizing the risk of hyperacute rejection when transplanted into humans. The modified pigs exhibited a significant reduction in xenoantigens, demonstrating the potential of CRISPR/Cas9 for creating viable xenograft donors (Tanihara et al., 2021).

 

Yue et al. (2020) conducted research involving the creation of pigs with multiple genetic modifications, including the inactivation of porcine endogenous retroviruses (PERVs) and the introduction of human transgenes to enhance immunological compatibility. These pigs showed normal physiology and improved resistance to human immune responses, highlighting the efficacy of CRISPR/Cas9 in generating complex, multi-gene modifications (Yue et al., 2020).

 

Obando et al. (2021) focused their research on modifying genes involved in immune tolerance. For instance, pigs engineered to express human leukocyte antigen-G (HLA-G) and other immune regulatory proteins have shown promise in reducing natural killer (NK) cell activity and promoting tolerance of xenografts (Obando et al., 2021).

 

5.3 Potential and limitations of CRISPR/Cas9 for improving graft survival

CRISPR/Cas9 technology allows for precise and efficient genetic modifications, enabling the elimination of immunogenic antigens and the introduction of human genes that can protect grafts from immune attack. This can significantly reduce the risk of hyperacute and acute rejection, potentially making xenotransplantation a viable solution to the organ shortage crisis (Cowan et al., 2019). Additionally, CRISPR/Cas9 can be used to deactivate PERVs, reducing the risk of zoonotic infections (Ross and Coates, 2018).

 

Despite its advantages, CRISPR/Cas9 has limitations, including the risk of off-target effects, where unintended parts of the genome are edited, potentially leading to unforeseen consequences. Additionally, achieving complete and uniform gene edits across all cells in an organ remains challenging, which can result in mosaicism. Ethical concerns regarding genetic modifications also need to be addressed before widespread clinical application (Naeimi Kararoudi et al., 2018).

 

While CRISPR/Cas9 technology holds great promise for improving the compatibility and survival of pig-to-human xenografts, further research and ethical considerations are essential to fully realize its potential in clinical settings.

 

6 Mechanisms of Genetic Modifications for Graft Survival

6.1 Mechanisms reducing hyperacute rejection

Hyperacute rejection is a significant barrier in xenotransplantation, primarily driven by preformed antibodies against pig antigens in the human recipient. Genetic modifications in pigs have been pivotal in mitigating this response. One of the most critical modifications is the knockout of the alpha-1,3-galactosyltransferase (GTKO) gene, which eliminates the expression of the alpha-Gal epitope, a major target for human antibodies. Studies have shown that kidneys from GTKO pigs transplanted into non-human primates (NHPs) and brain-dead human recipients did not exhibit signs of hyperacute rejection, indicating the effectiveness of this genetic modification (Firl and Markmann, 2022; Montgomery et al., 2022; Lei et al., 2022). Additionally, the insertion of human complement regulatory proteins, such as CD46 and CD55, further protects the xenograft from complement-mediated damage, which is a crucial component of hyperacute rejection (Lei et al., 2022; Goerlich et al., 2020).

 

6.2 Mechanisms mitigating acute vascular and cellular rejection

Acute vascular and cellular rejection occurs days to weeks post-transplantation and involves immune responses that target the endothelial cells of the graft. Genetic modifications to express human complement regulatory proteins (such as CD46, CD55, and CD59) and coagulation regulatory proteins (such as thrombomodulin) in pigs have shown to be effective in preventing complement activation and controlling coagulation, thereby protecting the graft from immune-mediated damage. The expression of these proteins helps maintain vascular integrity and reduces inflammatory responses within the graft (Burdorf et al., 2018; Singh et al., 2018).

 

Additionally, the expression of anti-inflammatory genes such as heme oxygenase-1 (HO-1) and human leukocyte antigen-G (HLA-G) in genetically modified pigs has been shown to mitigate acute cellular rejection. These genes help modulate the immune response, reduce inflammation, and promote immune tolerance (Obando et al., 2021; Ryczek et al., 2021).

 

6.3 Enhancing long-term graft survival through genetic stability and resistance to chronic rejection

Chronic rejection develops over months to years and is driven by both immune and non-immune mechanisms, including fibrosis and vascular occlusion. Genetic modifications that enhance the graft’s intrinsic resistance to immune-mediated damage and promote tissue repair are crucial for long-term graft survival. The introduction of genes encoding for anti-apoptotic proteins, such as Bcl-2, and anti-inflammatory cytokines, such as interleukin-10 (IL-10), can help in maintaining graft function and reducing chronic inflammation (Coe et al., 2020).

 

Furthermore, the incorporation of human CD47, which provides a "don't eat me" signal to macrophages, helps in preventing phagocytosis of the graft cells, thereby reducing chronic rejection. This modification, along with the expression of human regulatory proteins such as endothelial protein C receptor (EPCR) and tissue factor pathway inhibitor (TFPI), can significantly enhance the graft’s resistance to both immune attack and coagulation-related injuries (Burdorf et al., 2021; Lei et al., 2022).

 

These modifications not only help in reducing hyperacute, acute, and chronic rejection but also enhance the long-term stability and functionality of the grafts, making xenotransplantation a viable solution for addressing organ shortages.

 

7 Ethical and Regulatory Considerations

7.1 Ethical issues related to genetic engineering in xenotransplantation

Genetic engineering in xenotransplantation raises significant ethical concerns, particularly regarding the welfare and rights of the animals used as organ donors. The process of creating genetically modified pigs involves invasive procedures and often results in the animals being kept under conditions that fail to meet their biological and psychological needs. This raises questions about the ethical justification for using animals in this way, balancing the potential human benefits against the welfare of the animals (Eissa et al., 2022; Johnson, 2022). Moreover, there are broader ethical questions about the implications of genetic engineering, such as interfering with natural species boundaries and the long-term impact on both human and animal genetics (George, 2022).

 

7.2 Regulatory frameworks for genetic modifications in xenotransplantation

The regulatory landscape for xenotransplantation is complex and varies significantly between countries. Regulatory bodies such as the United States Food and Drug Administration (FDA) and the World Health Organization (WHO) have established guidelines to ensure the safety and ethical conduct of xenotransplantation research. These frameworks require rigorous preclinical testing to demonstrate the safety and efficacy of genetically modified organs and mandate continuous monitoring to prevent zoonotic disease transmission (Goerlich et al., 2019; Arcidiacono, 2020). Additionally, some countries have specific regulations addressing the ethical issues associated with the use of animals, ensuring that animal welfare is considered in the research protocols (Hawthorne and Cowan, 2020).

 

7.3 Public perception and acceptance of genetically modified xenografts

Public perception and acceptance of xenotransplantation are critical for its successful implementation. There are significant concerns about the safety of xenotransplantation, particularly the risk of cross-species disease transmission. Ethical concerns about animal welfare and the naturalness of using genetically modified animals also influence public opinion. To address these issues, it is essential to engage with the public through transparent communication about the risks and benefits of xenotransplantation and the measures taken to mitigate these risks (Entwistle et al., 2022; Gusmano, 2022). Educational initiatives and public consultations can help build trust and acceptance, ensuring that the ethical and societal implications are thoroughly considered and addressed.

 

In conclusion, while xenotransplantation holds great promise for addressing the organ shortage crisis, it raises complex ethical and regulatory challenges. Ensuring ethical conduct, robust regulatory oversight, and public engagement are crucial for the responsible advancement of xenotransplantation.

 

8 Case Studies and Clinical Trials

8.1 Analysis of successful case studies in pig-to-human xenotransplantation

Recent case studies have demonstrated significant progress in pig-to-human xenotransplantation, showcasing the feasibility and initial success of genetically modified pig organs functioning in human recipients.

 

A successful case involved the transplantation of porcine kidneys into a human decedent model, where the kidneys maintained vascular integrity and did not experience hyperacute rejection over a 74-hour period (Porrett et al., 2022).

 

Porrett et al. (2022) reported the first use of clinical-grade genetically modified pig kidneys for xenotransplantation in a human brain-dead model. The study utilized genetically edited pigs that had certain carbohydrate antigens and complement-mediated cytotoxic reactions removed to improve transplant outcomes. The transplanted pig kidneys survived for 74 hours within the recipients, with no observed hyperacute rejection reactions. The kidneys were able to produce urine post-transplantation, though the creatinine clearance rate did not recover. Additionally, kidney biopsies showed signs of thrombotic microangiopathy, which did not worsen. The study also monitored potential transmission of porcine-derived viruses, with no transmission of porcine endogenous retroviruses detected. Through this research, the authors demonstrated the feasibility of xenotransplantation in humans and highlighted key issues that require further investigation to enhance the safety and efficacy of xenotransplantation.

 

The histological sections in Figure 2 illustrate the adaptation process of pig kidneys in the human body and the changes in immune response following xenotransplantation. The initial stages (Figures 2A and 2B) display mild to moderate acute tubular damage in the kidneys, primarily due to cold ischemia. The glomeruli and capillary structures remain normal, indicating that the initial transplantation did not immediately trigger significant vascular or tissue damage. On the first day post-transplant (Figures 2C and 2D), significant congestion and thrombosis in the glomerular capillaries are evident, indicating the presence of thrombotic microangiopathy (TMA). The emergence of TMA may be related to the immune system's response to unique antigens in the pig kidney, although no deposition of antibodies or complement components was detected in this experiment. This reaction suggests that, despite genetic editing reducing antigenicity, the human immune system may still recognize and react to pig kidney cells.

 

By the third day post-transplant (Figures 2E and Figures 2F), although there is some improvement in glomerular lesions, the tubular damage has worsened to acute necrosis, likely related to hypoxia and ongoing immune attacks. The kidney's response may be influenced by various factors, including post-transplant management, the effectiveness of immunosuppressive treatments, and other unknown biological interactions. These pathological changes display the kidney's adaptation process when faced with challenges from the human immune system and also reveal the importance of optimizing immunosuppressive strategies in future clinical applications of xenotransplantation to minimize organ damage and improve graft survival.

 

Another study involving the transplantation of genetically engineered pig kidneys into a brain-dead human recipient showed no signs of hyperacute rejection. The kidneys produced urine and remained viable for 54 hours, highlighting the potential for these genetically modified organs to function in a human environment (Montgomery et al., 2022).

 

Montgomery et al. (2022) explored the potential of using genetically modified pigs as donors to address the human organ transplant demand. Due to acute rejection reactions caused by human antibodies to specific glycosylation changes, the pigs in the study were genetically edited to remove the alpha-1,3-galactosyltransferase gene to reduce immune rejection. The study involved transplanting pig kidneys into two brain-dead human recipients and observing them for 54 hours with ventilator support. Post-transplant, recipients immediately produced urine, and kidney function indicators, such as the glomerular filtration rate, improved. Biopsy examinations found no acute or antibody-mediated rejection responses. Additionally, the experiment included monitoring and prevention of potential porcine-derived diseases. This study demonstrated the short-term compatibility and functionality of genetically modified pig kidneys in human recipients, providing important data for future clinical applications.

 

Figure 3 displays the condition of pig kidneys transplanted into human recipients. Part A shows the immediate observation of the pig kidney post-transplant, where the kidney appears pink and healthy, with no obvious signs of ischemia or infarction. Part B presents the pig kidney 54 hours after transplantation, maintaining a pink color and healthy appearance. Parts C and D show the kidney conditions of the second recipient immediately after transplantation and 54 hours later, respectively, demonstrating a similarly good appearance. Part E depicts the setup connecting the ureter to the drainage system, specifically designed to collect urine produced by the pig kidney to assess its functionality. These images indicate that the pig kidneys were able to maintain good blood supply and functional status post-transplantation.

 

8.2 Ongoing and future clinical trials focusing on genetic determinants

Ongoing and future clinical trials are focusing on optimizing genetic modifications to improve graft survival and reduce rejection. Several trials are investigating the efficacy of triple-knockout (TKO) pigs, which lack the three major carbohydrate xenoantigens, combined with the expression of human complement regulatory proteins. These genetic modifications are crucial for reducing the antigenicity of pig organs and improving compatibility with human recipients (Cooper et al., 2019). Upcoming trials aim to test the long-term functionality and safety of these genetically engineered organs in human recipients, paving the way for broader clinical applications (Xu et al., 2022).

 

8.3 Outcomes and lessons learned from clinical applications

The outcomes of initial clinical applications of pig-to-human xenotransplantation have provided valuable insights into the challenges and potential of this technology. One significant lesson is the importance of selecting appropriate genetic modifications to address specific immunological barriers. For example, the expression of human thrombomodulin in genetically modified pigs has been shown to prevent intracardiac thrombus formation in pig-to-baboon cardiac xenotransplantation models, indicating its potential benefit in human trials (Goerlich et al., 2020). Additionally, the need for rigorous immunosuppressive protocols and continuous monitoring to prevent rejection and manage complications has been emphasized in these studies.

 

Overall, these case studies and clinical trials highlight the progress made in xenotransplantation and the critical role of genetic modifications in overcoming immunological challenges. Continued research and clinical testing will be essential to further refine these techniques and improve the long-term success of pig-to-human organ transplants.

 

9 Challenges and Future Directions

9.1 Technical challenges in achieving long-term graft survival

Achieving long-term graft survival in pig-to-human xenotransplantation involves addressing several technical challenges. One of the primary hurdles is overcoming hyperacute rejection, which can occur minutes to hours after transplantation due to the presence of preformed antibodies in the recipient that recognize pig antigens. Advanced genetic modifications, such as the knockout of the alpha-Gal gene, have been critical in reducing this risk. However, other forms of rejection, including acute vascular and cellular rejection, still pose significant challenges. These rejections occur due to complex immune responses involving complement activation, coagulation pathways, and cellular immunity, requiring a multifaceted approach to immunosuppression and genetic modification (Montgomery et al., 2022).

 

Another technical challenge is the risk of zoonotic infections. Porcine endogenous retroviruses (PERVs) can potentially be transmitted to human recipients. While genetic modifications to inactivate PERVs have been developed, ensuring the complete safety of xenotransplantation remains a priority (Xi et al., 2023).

 

9.2 Emerging trends and innovative approaches

Emerging trends and innovative approaches are continually evolving to address these challenges. CRISPR/Cas9 technology has revolutionized genetic engineering, allowing precise edits to the pig genome to knock out immunogenic genes and introduce human protective genes. This technology has facilitated the development of pigs with multiple genetic modifications aimed at reducing immunogenicity and enhancing compatibility with the human immune system (Zeng, 2023).

 

Furthermore, the use of chimeric organs, where human cells are introduced into developing pig organs, is an innovative approach to reducing rejection risks. This method aims to create organs that are more human-like in their cellular makeup, potentially improving graft acceptance and function (Cengiz and Wareham, 2019).

 

9.3 Interdisciplinary research and collaboration opportunities

The field of xenotransplantation benefits immensely from interdisciplinary research and collaboration. Collaboration between geneticists, immunologists, transplant surgeons, and bioethicists is essential for advancing the science and addressing the complex ethical and technical challenges. Interdisciplinary efforts have led to significant advancements, such as the development of genetically modified pigs with human-like immune tolerance, and have paved the way for potential clinical applications (Lu et al., 2020).

 

Future research will likely focus on fine-tuning genetic modifications to further reduce immunogenicity and enhance graft survival, as well as developing new immunosuppressive therapies tailored specifically for xenotransplantation. Collaborative networks and research consortia will play a crucial role in facilitating these advancements and ensuring the translation of preclinical findings into clinical practice (Niu et al., 2020).

 

In conclusion, while significant challenges remain, ongoing innovations and interdisciplinary collaborations hold promise for the successful implementation of pig-to-human xenotransplantation, potentially addressing the global shortage of human organs for transplantation.

 

10 Concluding Remarks

The systematic review on genetic determinants of long-term graft survival in pig-to-human xenotransplantation has highlighted several critical insights and findings. Key genetic modifications, such as the knockout of xenoantigens (e.g., GGTA1, CMAH, β4GalNT2) and the introduction of human complement regulatory proteins (e.g., CD46, CD55, thrombomodulin), have shown significant promise in reducing hyperacute and acute vascular rejection. Studies have demonstrated that genetically engineered pigs lacking major immunogenic antigens and expressing human regulatory genes can achieve improved graft survival and function. Furthermore, innovative approaches like CRISPR/Cas9-mediated genetic modifications and the use of chimeric organs have opened new avenues for enhancing graft compatibility and reducing immune rejection.

 

The findings from this study have significant implications for future research and clinical practice. Continued advancements in genetic engineering, particularly through the use of CRISPR/Cas9 technology, will be crucial in creating more refined and effective genetic modifications to improve graft survival. Future research should focus on long-term studies to assess the durability and functionality of genetically modified xenografts in human recipients. Additionally, the development of new immunosuppressive therapies tailored specifically for xenotransplantation is essential to manage immune responses and enhance graft longevity. Collaborative efforts across disciplines, including genetics, immunology, and transplantation surgery, will be vital in driving these advancements forward.

 

As the field of xenotransplantation advances, it is imperative to address the ethical considerations associated with genetic modifications and animal welfare. Researchers and policymakers must work together to establish ethical guidelines and regulatory frameworks that ensure the responsible and humane use of genetically modified animals. Public engagement and education are also critical to addressing societal concerns and building trust in the safety and efficacy of xenotransplantation. Continued exploration and innovation in this field hold the potential to address the global shortage of human organs for transplantation, offering hope to patients with end-stage organ failure. Therefore, a call to action is warranted for sustained research efforts, interdisciplinary collaboration, and ethical vigilance to realize the full potential of xenotransplantation.

 

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

Arcidiacono J., 2020, US food and drug administration regulatory approaches for xenotransplantation products and xenografts, Xenotransplantation, 27(6): e12626.

https://doi.org/10.1111/xen.12626

PMid:32776377

 

Burdorf L., Azimzadeh A., and Pierson R., 2018, Progress and challenges in lung xenotransplantation: an update, Current Opinion in Organ Transplantation, 23: 621-627.

https://doi.org/10.1097/MOT.0000000000000582

PMid:30234737

 

Cengiz N., and Wareham C., 2019, Pig-to-human xenotransplantation: Overcoming ethical obstacles, South African Journal of Bioethics and Law, 12(2): 66-71.

https://doi.org/10.7196/SAJBL.2019.v12i2.677

 

Coe T., Detelich D., Rickert C., Carroll C., Serifis N., Matheson R., Raigani S., Rosales I., Qin W., Kan Y., Layer J., Youd M., Westlin W., Kimura S., Azimzadeh A., Yang L., and Markmann J., 2020, Prolonged survival of genetically modified pig livers during machine perfusion with human blood, Transplantation, 104(S3): S37.

https://doi.org/10.1097/01.tp.0000698436.68163.75

 

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

 

Cooper D., Hara H., Iwase H., Yamamoto T., Wang Z., Jagdale A., Bikhet M., Nguyen H., Foote J., Paris W., Ayares D., Kumar V., Anderson D., Locke J., and Eckhoff D., 2020, Pig kidney xenotransplantation: progress toward clinical trials, Clinical Transplantation, 35(1): e14139.

https://doi.org/10.1111/ctr.14139

PMid:33131148

 

Cowan P., Hawthorne W., and Nottle M., 2019, Xenogeneic transplantation and tolerance in the era of CRISPR-Cas9, Current Opinion in Organ Transplantation, 24: 5-11.

https://doi.org/10.1097/MOT.0000000000000589

PMid:30480643

 

Cyprian W., 2020, Xenotransplantation of the pancreas: a reality check of its historical emergence, International Journal of Diabetes and Clinical Research, 46(6): 987-994.

 

Eissa N., Badrkhan S., Mohamed M., Shaban J., Shahban R., and Dawoud M, 2022, Xenotransplantation: past, present, and future directions, Highlights in BioScience, 5: 1-15.

https://doi.org/10.36462/H.BioSci.202205

 

Entwistle J., Sade R., and Drake D., 2022, Clinical xenotransplantation seems close: ethical issues persist, Artificial organs, 7(4): 1410131.

https://doi.org/10.1111/aor.14255

PMid:35451522

 

Firl D., and Markmann J., 2022, Measuring success in pig to non‐human‐primate renal xenotransplantation: systematic review and comparative outcomes analysis of 1051 life‐sustaining NHP renal allo‐ and xeno‐transplants, American Journal of Transplantation, 22: 1527-1536.

https://doi.org/10.1111/ajt.16994

PMid:35143091

 

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

 

George A., 2022, Ethics, virtues and xenotransplantation, Perfusion, 39(2): 334-343.

https://doi.org/10.1177/02676591221140767

PMid:36382884 PMCid:PMC10900854

 

Goerlich C., Chan J., and Mohiuddin M., 2019, Regulatory barriers to xenotransplantation, Current Opinion in Organ Transplantation, 24(5): 522-526

https://doi.org/10.1097/MOT.0000000000000678

PMid:31361629 PMCid:PMC10243300

 

Goerlich C., Kaczorowski D., Singh A., Abdullah M., Lewis B., Zhang T., Tatarov I., Sentz F., Hershfeld A., Odonkor P., Williams B., Strauss E., Ayares D., Griffith B., and Mohiuddin M., 2020, Human thrombomodulin transgene expression prevents intracardiac thrombus in life supporting pig-to-baboon cardiac xenotransplantation, The Journal of Heart and Lung Transplantation: the Official Publication of the International Society for Heart Transplantation, 39(4S): S145.

https://doi.org/10.1016/j.healun.2020.01.1069

 

Goerlich C., Singh A., Kaczorowski D., Abdullah M., Lewis B., Tatarov I., Hershfeld A., Zhang T., Strauss E., Odonkor P., Williams B., Ayares D., Griffith B., and Mohiuddin M., 2020, Lack of human thrombomodulin gene expression in life supporting pig-to-baboon cardiac xenotransplantation models accelerate antibody mediated rejection, Transplantation, 104(S3): S635.

https://doi.org/10.1097/01.tp.0000702028.97415.09

 

Gulyaev V., Khubutiya M., Novruzbekov M., Mironov A., Olisov O., Lutsyk K., Zhuravel S., Magomedov K., Akhmetshin R., and Yaremin B., 2019, Xenotransplantation: history, problems and development prospects, transplantologiya, The Russian Journal of Transplantation, 11: 1.

https://doi.org/10.23873/2074-0506-2019-11-1-37-54

 

Gusmano M., 2022, Xenotransplantation clinical trials and the need for community engagement, The Hastings Center report, 52(5): 42-43.

https://doi.org/10.1002/hast.1420

PMid:36226876

 

Hawthorne W., and Cowan P., 2020, Xenotransplantation in Australia: development of the regulatory process, Xenotransplantation, 27(3): e12603.

https://doi.org/10.1111/xen.12603

PMid:32506483

 

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

 

Hess N., and Kaczorowski D., 2023, The history of cardiac xenotransplantation: early attempts, major advances, and current progress, Frontiers in Transplantation, 2: 1125047.

https://doi.org/10.3389/frtra.2023.1125047

PMid:38993853 PMCid:PMC11235224

 

Johnson L., 2022, Existing ethical tensions in xenotransplantation, Cambridge Quarterly of Healthcare Ethics, 31: 355-367.

https://doi.org/10.1017/S0963180121001055

PMid:35659820

 

Kararoudi M., Hejazi S., Elmas E., Hellström M., Kararoudi M., Padma A., Lee D., and Dolatshad H., 2018, Clustered regularly interspaced short palindromic repeats/Cas9 gene editing technique in xenotransplantation, Frontiers in Immunology, 9: 1711.

https://doi.org/10.3389/fimmu.2018.01711

PMid:30233563 PMCid:PMC6134075

 

Kaur G., Wright K., Mital P., Hibler T., Miranda J., Thompson L., Halley K., and Dufour J., 2020, Neonatal pig sertoli cells survive xenotransplantation by creating an immune modulatory environment involving CD4 and CD8 regulatory T cells, Cell Transplantation, 29: 1-21.

https://doi.org/10.1177/0963689720947102

PMid:32841048 PMCid:PMC7564626

 

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

 

Loike J., and Kadish A., 2018, Ethical rejections of xenotransplantation?, EMBO Reports, 19: e46337.

https://doi.org/10.15252/embr.201846337

PMid:29991538 PMCid:PMC6073069

 

Lopez K., Cross-Najafi A., Farag K., Obando B., Thadasina D., Isidan A., Park Y., Zhang W., Ekser B., and Li P., 2022, Strategies to induce natural killer cell tolerance in xenotransplantation, Frontiers in Immunology, 13: 941880.

https://doi.org/10.3389/fimmu.2022.941880

PMid:36072599 PMCid:PMC9441937

 

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

 

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, New England Journal of Medicine, 386(20): 1889-1898.

https://doi.org/10.1056/NEJMoa2120238

PMid:35584156

 

Niu, D., Ma, X., Yuan, T., Niu, Y., Xu, Y., Sun, Z., Ping, Y., Li, W., Zhang, J., Wang, T., and Church, G, 2020, Porcine genome engineering for xenotransplantation, Advanced Drug Delivery Reviews, 168: 229-245.

https://doi.org/10.1016/j.addr.2020.04.001

PMid:32275950

Obando B., Cross-Najafi A., Lopez K., Thadasina D., Zhang W., Isidan A., Park Y., Campaña G., Li P., and Ekser B., 2021, Regulation of human NK cell activation by expression of HLA class I molecules in pig endothelial cells, Proceedings of IMPRS, 4: 1.

https://doi.org/10.18060/25715

 

Porrett P., Cheung M., Asiimwe R., Erman E., Fucile C., Liu S., Sun C., Hanumanthu V., Pal H., Wright E., Ghajar-Rahimi G., Epstein D., Orandi B., Kumar V., Anderson D., Greene M., Bell M., Yates S., Moore K., Lafontaine J., Killian J., Baker G., Perry J., Reed R., Little S., Rosenberg A., George J., and Locke J., 2023, Spatiotemporal immune atlas of the first clinical-grade, gene-edited pig-to-human kidney xenotransplant, Nature Communications Volume, 15: 3140.

https://doi.org/10.21203/rs.3.rs-2382345/v1

 

Porrett P., Orandi B., Kumar V., Houp J., Anderson D., Killian A., Hauptfeld-Dolejsek V., Martin D., Macedon S., Budd N., Stegner K., Dandro A., Kokkinaki M., Kuravi K., Reed R., Fatima H., Killian J., Baker G., Perry J., Wright E., Cheung M., Erman E., Kraebber K., Gamblin T., Guy L., George J., Ayares D., and Locke J., 2022, First clinical‐grade porcine kidney xenotransplant using a human decedent model, American Journal of Transplantation, 22(4): 1037-1053.

https://doi.org/10.1111/ajt.16930

PMid:35049121

 

Reichart B., Längin M., Radan J., Mokelke M., Buttgereit I., Ying J., Fresch A., Mayr T., Issl L., Buchholz S., Michel S., Ellgass R., Mihalj M., Egerer S., Baehr A., Kessler B., Kemter E., Kurome M., Zakhartchenko V., Steen S., Sjöberg T., Paskevicius A., Krüger L., Fiebig U., Denner J., Godehardt A., Tönjes R., Milusev A., Rieben R., Sfriso R., Walz C., Kirchner T., Ayares D., Lampe K., Schönmann U., Hagl C., Wolf E., Klymiuk N., Abicht J., and Brenner P., 2020, Pig-to-non-human primate heart transplantation: The final step toward clinical xenotransplantation?, The Journal of Heart and Lung Transplantation: the Official Publication of the International Society for Heart Transplantation, 39(8): 751-757.

https://doi.org/10.1016/j.healun.2020.05.004

PMid:32527674

 

Ross M., Coates P., and Coates P., 2018, Using CRISPR to inactivate endogenous retroviruses in pigs: an important step toward safe xenotransplantation? Kidney international, 93(1): 4-6.

https://doi.org/10.1016/j.kint.2017.11.004

PMid:29198467

 

Ryczek, N., Hryhorowicz, M., Zeyland, J., Lipinski, D., and Słomski, R, 2021, CRISPR/Cas Technology in pig-to-human xenotransplantation research, International Journal of Molecular Sciences, 22(6): 3196.

https://doi.org/10.3390/ijms22063196

PMid:33801123 PMCid:PMC8004187

 

Singh A., Chan J., DiChiacchio L., Hardy N., Corcoran P., Lewis B., Thomas M., Burke A., Ayares D., Horvath K., and Mohiuddin M., 2018, Cardiac xenografts show reduced survival in the absence of transgenic human thrombomodulin expression in donor pigs, Xenotransplantation, 26(2): e12465.

https://doi.org/10.1111/xen.12465

PMid:30290025 PMCid:PMC6450784

 

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

 

Tatapudi V., and Griesemer A., 2022, Physiologic considerations of pig-to-human kidney xenotransplantation, Current Opinion in Nephrology and Hypertension, 32: 193-198.

https://doi.org/10.1097/MNH.0000000000000858

PMid:36683545

 

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, Journal of Biomedical Research, 33(4): 235-243.

https://doi.org/10.7555/JBR.32.20180018

PMid:30007952 PMCid:PMC6813527

 

Wu H., Lian M., and Lai L., 2023, Multiple gene modifications of pigs for overcoming obstacles of xenotransplantation, National Science Open, 2(5): 20230030.

https://doi.org/10.1360/nso/20230030

 

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 H., Zhang X., and Dou K., 2022, Key issues related to clinical trials of xenotransplantation, Chinese Journal of surgery, 60(10): 888-893 .

 

Yamada K., Ariyoshi Y., Pomposelli T., and Sekijima M., 2020, Co-transplantation of vascularized thymic graft with kidney in pig-to-nonhuman primates for the induction of tolerance across xenogeneic barriers, Methods in Molecular Biology, 2110: 151-171.

https://doi.org/10.1007/978-1-0716-0255-3_11

PMid:32002908 PMCid:PMC7366292

 

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

 

Yue Y., Xu W., Kan Y., Zhao H., Zhou Y., Song X., Wu J., Xiong J., Goswami D., Yang M., Lamriben L., Xu M., Zhang Q., Luo Y., Guo J., Mao S., Jiao D., Nguyen T., Li Z., Layer J., Li M., Paragas V., Youd M., Sun Z., Ding Y., Wang W., Dou H., Song L., Wang X., Le L., Fang X., George H., Anand R., Wang S., Westlin W., Güell M., Markmann J., Qin W., Gao Y., Wei H., Church G., and Yang L., 2020, Extensive germline genome engineering in pigs, Nature Biomedical Engineering, 5: 134-143.

https://doi.org/10.1038/s41551-020-00613-9

PMid:32958897

 

Zeng Y, 2023, CRISPR/Cas-mediated genome editing and its application on pig-to-human xenotransplantation, International Conference on Modern Medicine and Global Health (ICMMGH 2023), 127891: 1-7.

https://doi.org/10.1117/12.3000215

International Journal of Molecular Medical Science
• Volume 14
View Options
. PDF
. FPDF(win)
. FPDF(mac)
. HTML
. Online fPDF
Associated material
. Readers' comments
Other articles by authors
. Jessi J. White
Related articles
. Xenotransplantation
. Genetic modifications
. Graft survival
. Immunosuppression
. Pig-to-human transplantation
. Gene editing
Tools
. Post a comment