You are on page 1of 9

1226849

review-article20242024
CLLXXX10.1177/09636897241226849Cell TransplantationZhou et al

Review

Cell Transplantation

Mitigating Cross-Species Viral Infections


Volume 33: 1­–9
© The Author(s) 2024
Article reuse guidelines:
in Xenotransplantation: Progress, sagepub.com/journals-permissions
DOI: 10.1177/09636897241226849
https://doi.org/10.1177/09636897241226849

Strategies, and Clinical Outlook journals.sagepub.com/home/cll

Yenong Zhou1 , Shuyu Zhou2, Qian Wang3, and


Bing Zhang1

Abstract
Xenotransplantation holds great promise as a solution to address the critical shortage of organs, but it raises concerns
regarding the potential transmission of porcine viruses to recipients, leading to infections and even zoonotic diseases. Data
used in this review were mainly from literature of Pubmed database. Keywords included xenotransplantation, infection, virus,
and epidemiology. The original articles and critical reviews selected were relevant to this review’s theme. We review the
major viral infections of concern in xenotransplantation, their risk of transmission, diagnosis, treatment, and ways to prevent
infection. Then, we pivot to a comprehensive overview of the current status of xenotransplantation. In addition, we offer our
own insights and recommendations for propelling xenotransplantation forward, transitioning from preclinical experiments to
the critical phase of clinical trials. Viral infections pose considerable safety concerns within xenotransplantation, particularly
with the possibility of emerging or currently unidentified viruses. Clinical trials serve as a crucial platform to progress the
safety standards of xenotransplantation. However, further studies and dedicated efforts are required to effectively translate
findings into practical applications that can improve safety measures in this field.

Keywords
xenotransplantation, infection, virus and epidemiology

Introduction these infections and their pathogenic effects may be even


more pronounced, given the imperative need for immuno-
A persistent worldwide shortage of organs from deceased suppression in xenotransplantation11.
human donors for transplantation plagues patients with end-
stage organ failure1. For instance, a survey by the International
Society for Heart and Lung Transplantation (ISHLT) high- Viruses
lights the stark reality that a vast number of end-stage heart Drawing from experiences in allotransplantation and insights
failure patients eagerly await a heart transplant, with fewer from preclinical models, viral infections loom as our fore-
than 4% fortunate enough to receive one2. Xenotransplantation, most concern, with the potential to even trigger zoonotic
the transfer of living cells, tissues, or organs from genetically transmissions. This concern finds stark confirmation in the
engineered pigs, emerges as a promising alternative to allevi-
ate this organ shortage crisis3–5. In 2022, a historic milestone
1
was achieved at the University of Maryland Medical Center,  epartment of Cardiovascular Surgery, Xijing Hospital, Air Force
D
where the first-ever gene-edited pig heart was successfully Medical University, Xi’an, China
2
Inner Mongolia Autonomous Region Hospital of Traditional Chinese
transplanted into a human recipient, yielding encouraging Medicine, Hohhot, China
data6. This remarkable achievement has significantly bol- 3
Nutriology Department, Qingdao Special Servicemen Recuperation
stered the confidence of scientists in the field. Nevertheless, Center of PLA Navy, Qingdao, China
the successful application of xenografts carries an inherent Submitted: September 24, 2023. Revised: January 2, 2024.
risk—viral infection7. Infections can have dire conse- Accepted: January 3, 2024.
quences, impacting survival rates, exacerbating inflam­
Corresponding Author:
matory responses, and triggering immune rejection in Bing Zhang, Department of Cardiovascular Surgery, Xijing Hospital, Air
allotransplantation8–10. Anticipating the prevalence of infec- Force Medical University, 127 Changle West Road, Xi’an 710032, China.
tions in xenotransplantation, it is crucial to recognize that Email: 15771793132@163.com

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-
NonCommercial 4.0 License (https://creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and
distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages
(https://us.sagepub.com/en-us/nam/open-access-at-sage).
2 Cell Transplantation

case of the recipient of the world’s first pig heart xenotrans- yield longer amplicons, enhancing our ability to sequence and
plantation, whose unfortunate demise was suspected to result classify the detected PCMVs effectively25. However, it is
from a porcine cytomegalovirus (PCMV) infection12. This important to note that certain PCR methods may occasionally
sobering event underscores the inherent perils of viral infec- yield false negatives26. Therefore, there is a need for the adop-
tions in xenotransplantation. Following the guidelines out- tion of more sensitive PCR techniques. New diagnostic
lined by the American Society of Transplantation, we focus approaches, such as nested PCR and duplex real-time PCR
our analysis on three key viruses: PCMV, porcine lympho- systems, have been developed with improved parameters,
tropic herpesvirus (PLHV), and porcine endogenous retrovi- significantly bolstering their sensitivity and accuracy25,27. In
rus (PERV)13. Our investigation spans a comprehensive addition to PCR-based methods, immunological techniques
exploration of these viruses, encompassing their epidemiol- for PCMV detection have also seen advancements. Using
ogy, modes of transmission, monitoring techniques, and recombinant proteins corresponding to the two domains of
more. PCMV glycoprotein gB as antigens, researchers have
employed Western blot technology to analyze the presence of
PCMV-specific antibodies28. Furthermore, an indirect-block-
Porcine Cytomegalovirus ing enzyme-linked immunosorbent assay (ELISA) method
PCMV, officially known as suid beta herpesvirus 2 (SuBHV2) designed to detect the gB epitope has demonstrated impres-
in accordance with the classification by the International sive specificity at 98% and remarkable sensitivity at 97.8%29.
Committee on Taxonomy of Viruses (ICTV), belongs to the In terms of the detection timeframe, real-time PCR offers a
Roseolovirus genus, signifying its place within the Beta her- distinct advantage, as it facilitates the early detection of
pesvirinae subfamily14,15. Analysis of its genome sequence PCMV presence in piglets compared to antibody testing12,26.
has revealed a total length of 128,367 base pairs, encompass-
ing 79 predicted open reading frames (ORFs)14. PCMV
Porcine Lymphotropic Herpesvirus
exhibits a widespread presence among pig populations
worldwide. For instance, in the Hunan and Sichuan prov- PLHV-1, PLHV-2, and PLHV-3 belong to the gamma her-
inces of China, the prevalence of PCMV-positive pigs stands pesvirus family and are prevalent among pigs. A survey con-
at 96.4% and 84.4%, respectively16,17. PCMV has been ducted in a densely populated pig area in Northern Italy
linked to various ailments in pigs, including pneumonia, rhi- revealed the widespread presence of PLHVs, with preva-
nitis, small body size, miscarriages in female pigs, and edema lence rates of 28.97%, 10.79%, and 4.54% for PLHV-1,
of the heart and other organs18. Notably, PCMV has been PLHV-2, and PLHV-3, respectively. These viruses were not
associated with consumptive coagulopathy in a recent only detected in various pig tissues but were also associated
study19. In the context of xenotransplantation, PCMV infec- with specific clinical conditions30. PLHV confirmed to infect
tions have proven detrimental, leading to a reduction in graft persistently porcine B cell line L2331,32. While PLHV is gen-
survival time. Previous investigations have demonstrated erally benign in its natural host, it can pose a significant
that the presence of PCMV during porcine heart transplanta- health threat when transmitted to other species, causing
tion in baboons resulted in a significant decrease in survival severe diseases33. Surprisingly, no direct link between PLHV
times, with durations dwindling from 33 days to 20 days and and swine diseases has been established thus far. However,
from 195 days to 30 days, respectively20,21. Moreover, in research has indicated that PLHV-1 can induce posttrans-
2022, a groundbreaking xenotransplantation at the University plant lymphoproliferative disease (PTLD) in minipigs34,35.
of Maryland in Baltimore involved a pig heart transplant into The primary mode of PLHV transmission is horizontal, but
a human patient. This case raised concerns that PCMV may there is also a risk of vertical transmission from parent to
have contributed to the patient’s unfortunate demise, given offspring33,36. Currently, no treatments or vaccines are avail-
that the virus potentially entered the recipient’s body along able for these three PLHVs.
with the transplanted organ12. Similar to other herpesviruses Even strategies like early weaning and colostrum feeding
known for their resilience within host organisms, PCMV can have proven ineffective in eliminating PLHV37. Although
undergo reactivation in vivo under conditions of stress22. The practices such as cesarean section and barrier maintenance
study had reported the presence of antibody cross-reactivity have reduced PLHV infection rates from 80% to as low as
between PCMV and human herpesvirus 6 (HHV-6)23. While 3% to 12.8%, they are not foolproof38, necessitating the
PCMV is unlikely to infect human cells or replicate within development of sensitive detection methods and novel
the human body, uncertainties persist regarding its potential approaches for PLHV elimination.
to induce diseases akin to HHV-6, like roseola infantum, or PLHV presence has been systematically examined and
reduce the survival time of the pig transplant24. These find- quantified using PCR assays, with a particular focus on spe-
ings underscore the critical significance of PCMV in the con- cific real-time PCR techniques30,39. These assays have strate-
text of xenograft survival. gically employed primers and probes targeting key regions
Numerous polymerase chain reaction (PCR) methods are within the DNA polymerase gene and the gene responsible
at our disposal for the detection of PCMV, some of which for encoding glycoprotein B33. In addition to PCR-based
Zhou et al 3

approaches, the detection of antibody responses to recombi- Given the significant variation in PERV copy numbers
nant glycoprotein B of PLHV-1 has been explored using both across different pig breeds, employing PCR to quantify
Western blot assays and ELISA methods40,41. These compre- PERV copy numbers in various pig breeds becomes invalu-
hensive techniques have significantly contributed to our able for selecting the most suitable candidates for xenotrans-
understanding of PLHV dynamics and its impact on host plantation44,58. Among the available techniques, droplet
organisms. digital PCR (ddPCR) stands out as the most commonly used
method44. Furthermore, the presence of PERV particles can
be visually confirmed through electron microscopy, provid-
Porcine Endogenous Retroviruses ing an additional means of detection59. An innovative
Numerous viruses have been identified in both pigs and approach proposed in a study is the immunoperoxidase assay
humans, with PERVs standing out as particularly noteworthy. (IPA), designed to detect viral proteins in infected cells and
PERVs are known to be transmissible from pig cells to human antibodies against PERV in the serum of the infected host60.
cells, making them a subject of significant concern42. There These diverse methodologies offer essential tools for assess-
are three types of PERVs: PERV-A and PERV-B, which are ing and managing the risk associated with PERVs in xeno-
integrated into the genome of all pigs, and PERV-C, which is transplantation settings.
present in most, but not all, pigs. While PERV-A and PERV-B
are polytropic and capable of infecting various human cell
types, PERV-C is ecotropic and exclusively infects porcine Elimination of Porcine Viruses
cells. Intriguingly, PERV-A and PERV-C can combine to form
PERV-A/C recombinants. These recombinants exhibit a simi-
Control of Pig Donors
lar ability to infect human cells as PERV-A but have been Selection of pig herds. Notably, not all pigs harbor specific
shown to have a higher replication rate than PERV-A alone43. pathogens, underscoring the critical need to identify and
PCR amplification technology has proven effective in detect- assess the pathogens present in pigs for the purpose of select-
ing PERVs in the genomes of four major miniature pig breeds ing suitable donors in xenotransplantation endeavors. Estab-
in China44. Over time, PERVs have been observed to remain lishing an “exclusion list” serves as a fundamental foundation
active within their host organisms, with the number of copies for the screening of potential pig donors, and it necessitates
increasing 45. Notably, PERVs carry the potential to induce periodic revision in response to evolving global swine infec-
various health issues, including the development of tumors, tion epidemiology and evolving clinical experience7. As an
leukemias, and neurodegenerative diseases46,47. Recent illustrative example, the selection of pigs with diminished
research has also revealed that PERVs can stimulate the pro- expression of PERV-A and PERV-B or those lacking PERV-
duction of the pro-inflammatory chemokine CXCL10 in C can be a proactive measure to prevent the binding of
human monocytes and monocyte-derived primary cells, PERV-A and PERV-C61. Such negative pigs represent viable
thereby enhancing the innate immune response within the candidates for direct use in xenotransplantation procedures,
host48. To date, no transmission of PERVs has been observed thus mitigating the risk associated with PERV transmission.
in clinical trials or preclinical trials49,50. There are several pos-
sible explanations for this absence of transmission. It is con- Medicines. In situations where negative pigs are not readily
ceivable that PERVs are not released from the graft, or they available, an alternative approach involves the selection of
may be suppressed by intracellular restriction factors and the pigs with low viral loads, followed by treatment using vac-
innate immune response within the recipient51. cination or antiviral drugs62. Specifically, PCMV can be
The patients selected for the clinical porcine islet xeno- effectively inhibited by various antiviral drugs, including
transplantation were nonimmunocompromised individuals ganciclovir (a synthetic analogue of 20-deoxy-guanosine),
with diabetes. This characteristic might have contributed to cidofovir (employed in the treatment of HCMV-induced reti-
preventing the spread of PERV50. It has been demonstrated nitis in humans), foscarnet (functioning as a structural mimic
that PERVs can infect human cells52,53 and integrate into of the anion pyrophosphate, selectively inhibiting the pyro-
human genome in cell culture54. This raises concerns regard- phosphate binding site on viral DNA polymerase), acyclovir
ing the possibility of PERVs being transmitted through a (a guanosine analogue), and valaciclovir (a prodrug of acy-
transplant patient and potentially becoming infectious to clovir)63. Studies have demonstrated that both ganciclovir
individuals in contact with that patient, posing a potential and cidofovir exhibit superior efficacy in inhibiting PCMV
source for a widespread epidemic13,55. In addition, microchi- replication when compared to foscarnet and acyclovir64. It is
merism, which can be challenging to distinguish from PERV worth noting that as of now, there have been no reports of
infection, has been reported in the initial human xenograft successful anti-PCMV vaccines. Conversely, for PERV,
trials and in most preclinical trials involving nonhuman pri- inhibitors targeting viral reverse transcriptase and integrase
mates56,57. These findings underscore the critical importance have shown effectiveness. Among these, azidothymidine
of continuous monitoring and accurate identification of (AZT) stands out as the most potent inhibitor of reverse tran-
PERVs in xenotransplantation research. scriptase, while integrase inhibitors have demonstrated
4 Cell Transplantation

significant efficacy against PERV65–67. Effective neutralizing continuous inhibition of PERV replication76–78. PERV-
antibodies can be induced when different animal species are specific shRNA has demonstrated the ability to reduce
immunized with PERV’s recombinant transmembrane enve- PERV expression both in vitro (within PERV-producing
lope protein p15E and envelope glycoprotein gB7068. These human cells) and in vivo (in transgenic pigs engineered to
antibodies recognize an epitope in the p15E fusion peptide express PERV-specific shRNA)79. Such interventions con-
proximal region (FPPR), termed E1, and an epitope in the tribute to enhancing the safety of xenotransplantation proce-
membrane proximal external region (MPER), termed E269,70. dures. In addition, some research findings suggest that
Nonetheless, it is imperative to conduct extensive analyses certain immunosuppressant agents can reduce PERV expres-
using diverse animal models to comprehensively evaluate sion in vitro without synergistic or antagonistic effects on
the impact and effectiveness of PERV vaccines. RNAi-mediated PERV suppression80.

Cesarean delivery, colostrum deprivation, and early weaning.


Gene Editing
When negative animals are not available and vaccines or
antiviral drugs prove ineffective against viruses with low Gene editing stands out as an ideal approach for inactivating
viral loads, there are alternative methods for virus elimina- viruses embedded within the genome. Technologies such as
tion. Some viruses are transmitted vertically, highlighting the Zinc finger nuclease (ZFN) or clustered regularly interspaced
importance of preventing the vertical transmission of viruses short palindromic repeats–associated RNA-guided DNA
from sows to piglets. Studies have confirmed that practices endonuclease Cas9 (CRISPR/Cas9) can be harnessed to deac-
such as early weaning, colostrum deprivation, and cesarean tivate the PERV gene81. ZFN technology, designed to target
delivery can effectively eliminate the PCMV virus in multiple proviral sequences of PERV, exhibited high expres-
pigs37,71. However, it is important to note that these methods sion within the nucleus and effective interactions. However,
do not result in complete clearance of the PLHV virus, as its implementation induced extreme cytotoxicity in PERV-
experimental results have indicated the continued presence infected cells82. In contrast, CRISPR/Cas9 has shown greater
of PLHV viral DNA in early weaned pigs36,37. success. In 2015, Yang and her research team employed
CRISPR/Cas9 to eliminate 62 PERV copies in porcine kidney
Biosecurity of pig farms. Once the virus is successfully elimi- epithelial cell line (PK15), resulting in a reduction of PERV
nated, it is imperative to maintain the animal in strict isolation transmission to humans by more than 1,000 times53.
to prevent de novo infections or re-entry of the virus. For Subsequently, in 2017, Yang and her team used CRISPR-
instance, certain viruses like Hepatitis E virus (HEV) can per- Cas9 to inactivate all PERV instances in pig primary cell lines
sist in drinking water, feces, and buildings72,73. Therefore, and generated PERV-inactivated pigs through somatic cell
donor pigs should be raised and kept in biosecure facilities nuclear transfer42. Furthermore, in 2021, the team demon-
that isolate them from the external environment. These facili- strated that CRISPR-Cas9 and transposon technology could
ties should provide filtered air, sterilized water, and irradiated be used to engineer pigs with inactivated PERV, eliminating
food certified to be free of any mammalian protein. All mate- three xenoantigens and enabling the expression of nine human
rials entering these facilities must undergo autoclaving, and transgenes. This advancement enhanced the pig’s immune
personnel should enter through showers and wear specialized compatibility and coagulation compatibility with humans,
clothing74. Only through rigorous supervision and control of successfully addressing the issue of PERV safety in xeno-
all aspects can donor pigs attain the status of designated transplantation83. However, a notable challenge persists in the
pathogen-free (DPF). form of a highly sensitive method for measuring PERV copy
numbers and expression levels45,53,82. Recent studies have
explored the use of cytosine base editors (CBEs), which do
RNA Interference not induce DNA double-strand breaks (DSBs) like CRISPR-
RNA interference (RNAi) represents a swift and efficient Cas9, offering a means to edit PERV with reduced cytotoxic
method for gene expression suppression. This process effects. In addition, the plasmids employed for PERV editing
involves two primary steps: initially, double-stranded RNA are not integrated into the host genome, and they do not
(dsRNA) is cleaved into small interfering RNAs (siRNAs) impact the karyotype of modified cells84.
through the activity of bacterial ribonuclease III (RNase
III)-like enzymes. Subsequently, these siRNAs associate Current Status and Prospects for
with the RNA-induced silencing complex (RISC) and facili-
tate the degradation of target messenger ribonucleic acid
Clinical Trails
(mRNA)26,75. In the context of PERV, its expression can be Over the past decade, nonhuman primate models have wit-
significantly reduced through the use of siRNA molecules nessed remarkable advancements, thanks to the emergence
that correspond to various segments of viral genes such as of gene editing technologies and refined immunosuppres-
gag, pol, and env. The most potent sequences are carefully sive regimens. These innovations have propelled research to
selected and expressed as short hairpin RNA (shRNA) using new heights, as evidenced by documented achievements in
the polymerase III vector system. This approach enables the xenotransplantation outcomes. Researchers have reported
Zhou et al 5

instances of remarkable survival in pig-baboon heterotopic pathological changes. The adverse effects of brain death on
heart xenografts, with some cases extending up to an impres- myocardial function, marked by substantial anaerobic meta-
sive 945 days, and in orthotopic xenografts, survival periods bolic and hemodynamic decline, trigger a catecholamine
of up to 195 days have been achieved85–87. In a groundbreak- storm resulting in heightened tachycardia and hypertension.
ing development, the University of Maryland School of This escalation drives increased cardiac output and myocar-
Medicine disclosed the successful transplantation of gene- dial oxygen consumption, worsening underlying myocardial
edited pig hearts into baboons. These modified pig hearts, ischemia, and significantly elevating the risk of postopera-
with six genes edited, exhibited remarkable resilience, tive transplant failure97–100. But xenotransplantation studies
surviving in baboons for a remarkable 264 days with the from recently deceased donors are critical to gathering the
assistance of life support88. Furthermore, in a significant additional human data needed to advance the field. Several
milestone in 2022, scientists achieved the transplantation of crucial steps must be completed before FDA approval can be
gene-edited pig hearts into two recently deceased humans. obtained, including the testing of a clinical immunosuppres-
Notably, there were no early signs of rejection observed in sion model, ensuring the genetic modifications of xenografts
either transplanted organ. The transplanted hearts func- are appropriate, and establishing a robust viral surveillance
tioned normally with the application of standard post­ protocol with confirmation of biocompatibility101–104. Several
transplant medications, obviating the need for additional institutions have announced their intentions to initiate clini-
mechanical support89. These groundbreaking successes cal trials in kidney xenotransplantation, with a phase I clini-
underscore the promising prospects of xenotransplantation cal trial of gene-edited pig-human kidney transplantation
as a viable solution to address the critical shortage of human registered in 2023, pending U.S. FDA authorization105.
donor organs. The successes in kidney xenotransplantation Despite the substantial progress made, the absolute risk of
have also been nothing short of impressive, with the longest infection in xenotransplantation remains uncertain. Early clin-
recorded survival period extending to an astounding 499 ical trials will provide valuable insights into the most suitable
days90. Notably, in 2021, several scientific research teams monitoring and prophylaxis strategies for xenotransplantation.
achieved significant milestones by transplanting gene- Given the immunosuppressed state of organ recipients, the
edited pig kidneys into two brain-dead human recipients. anticipation of infections is crucial. Conducting routine pre-
Remarkably, no instances of hyperacute rejection were transplant screenings for recipients will help identify latent
observed in these groundbreaking procedures91,92. In addi- infections that necessitate ongoing surveillance or the imple-
tion, substantial strides have been made in the field of liver mentation of prophylactic therapies. Screening source animals
xenotransplantation. In a noteworthy development in 2020, for latent and active infections using available assays can help
13 gene-edited pig-rhesus monkeys demonstrated remark- restrict donor-derived infections to a certain extent. Regular
able resilience, surviving for 26 days following heterotopic monitoring following FDA and other relevant guidance docu-
auxiliary liver transplantation93. Building on this progress, ments involves employing microbe-specific assays. In addi-
in 2023, a gene-edited pig-macaque orthotopic auxiliary tion, the implementation of advanced unbiased metagenomic
liver transplant achieved a survival period of 34 days, sequencing methods can aid in surveillance for both known
marking another significant advancement in the field94. In and unknown organisms106,107. Advancements in microbiol-
our center, we successfully conducted multiorgan xeno- ogy, such as quantitative molecular assays for viruses and
transplantation procedures in both 2020 and 2022, marking unbiased metagenomic sequencing, enable the screening and
significant milestones in the field of xenotransplantation95,96. monitoring of recipients and donors for infections, even in the
Notably, in 2022, we achieved a remarkable feat by trans- absence of symptoms. But these methods have not yet been
planting three organs (liver, kidney, and heart) and three tis- validated or approved for clinical use and are known to incur
sues (cornea, skin, and bones) from a 6-gene edited pig into high costs108. Blood samples collected from recipients and
four rhesus monkeys. In these groundbreaking procedures, contacts can be systematically archived at standard intervals.
the heterotopic heart transplant recipients exhibited a sur- This archival practice serves the purpose of facilitating future
vival period of 20 days, while the monkey receiving a com- epidemiological studies or advancements in unbiased metage-
bined liver and kidney transplant survived for 14 days96. nomic sequencing techniques. Xenotransplantation recipients
While the successful outcomes of preclinical experiments displaying signs of infection, such as fever, hypotension, or
in recent years have fueled optimism about the progression graft dysfunction, may undergo various diagnostic procedures.
of xenotransplantation to clinical applications, it is important These include blood, urine, and/or sputum cultures, alongside
to note that the Food and Drug Administration (FDA) has not relevant radiological examinations and invasive diagnostics
yet granted approval for clinical trials. The world’s first pig involving microbiological and histopathological analyses109.
heart transplant was conducted under the FDA’s “compas- Implementing stringent infection control measures is crucial
sionate use” provision, which allows for experimental treat- throughout this process. We believe that as clinical data con-
ments when a patient is facing a serious or life-threatening tinue to emerge, regulatory, and control guidance on infection
medical condition6. Using deceased models for xenotrans- will evolve accordingly.
plantation research also poses certain limitations. Studies Based on our experiences, we offer the following recom-
show hearts procured after brain death manifest distinct mendations for future clinical trials: First, involve virologists
6 Cell Transplantation

in the research design phase to provide valuable insights62. ORCID iD


Second, given the presence of numerous new or unfamiliar Yenong Zhou https://orcid.org/0009-0006-5727-301X
viruses in donor animals, the development of more sensitive
detection and elimination methods is imperative26. Third, References
considering that the use of immunosuppressants in xenograft
1. Cooper DKC, Gaston R, Eckhoff D, Ladowski J, Yamamoto
recipients can exacerbate infection symptoms, it is essential to T, Wang L, Iwase H, Hara H, Tector M, Tector AJ.
assess and optimize the recipient’s condition prior to surgery. Xenotransplantation-the current status and prospects. Br Med
Finally, implementing meticulous infection control measures Bull. 2018;125(1):5–14.
tailored to various groups such as xenotransplant recipients, 2. Hsich E, Singh TP, Cherikh WS, Harhay MO, Hayes D,
healthcare workers, contacts, and others is essential. Perch M, Potena L, Sadavarte A, Lindblad K, Zuckermann
A, Stehlik J, et al. The International thoracic organ trans-
plant registry of the International Society for Heart and
Conclusions Lung Transplantation: thirty-ninth adult heart transplantation
Recent discoveries have underscored the paramount impor- report-2022; focus on transplant for restrictive heart disease. J
tance of viral safety in ensuring the success of xenotrans- Heart Lung Transplant. 2022;41(10):1366–75.
3. Food and Drug Administration. Guidance for industry: precau-
plantation. In recent years, significant strides have been
tionary measures to reduce the possible risk of transmission of
made in developing highly sensitive and specific methods zoonoses by blood and blood products from xenotransplanta-
for detecting swine viruses, effectively eliminating the tion product recipients and their contacts. 2002. https://www.
majority of known viral threats. Nevertheless, the ongoing federalregister.gov/documents/2002/02/11/02-3200/draft-
emergence of new and latent viruses poses an ongoing chal- guidance-for-industry-precautionary-measures-to-reduce-the-
lenge to the safety of xenotransplantation, as these viruses possible-risk-of-transmission-of.
carry the potential to infect humans or even trigger fresh out- 4. Garry DJ, Weiner JI, Greising SM, Garry MG, Sachs DH.
breaks of disease in pig populations. Looking ahead, our ulti- Mechanisms and strategies to promote cardiac xenotransplan-
mate aspiration is to comprehensively address and resolve tation. J Mol Cell Cardiol. 2022;172:109–19.
the issue of microbiological safety in xenotransplantation. 5. Brown JL, Voth JP, Person K, Low WC. A technological
This ambitious goal will require ongoing vigilance, research, and regulatory review on human-animal chimera research: the
current landscape of biology, law, and public opinion. Cell
and the continuous development of cutting-edge strategies to
Transplant. 2023;32:1–18.
safeguard the health of both recipients and donor animals. 6. Mohiuddin MM, Singh AK, Scobie L, Goerlich CE, Grazioli
A, Saharia K, Crossan C, Burke A, Drachenberg C, Oguz C,
Authors’ Contributions Zhang T, et al. Graft dysfunction in compassionate use of
YZ, SZ, and QW worked together to supply a dynamic perspective genetically engineered pig-to-human cardiac xenotransplan-
of xenotransplantation in this review article. BZ provided critical tation: a case report. Lancet. 2023;402:397–410.
feedback and revised the article. YZ, SZ, and QW contributed 7. Fishman JA. Infectious disease risks in xenotransplantation.
equally to this work. Am J Transplant. 2018;18(8):1857–64.
8. Shultes KC, Shuster JE, Micek S, Vader JM, Balsara K, Itoh
Ethical Approval A, Tellor BR. Outcomes and predictors of early infection
after heart transplantation. Surg Infect. 2018;19(5):516–22.
This study was approved by our institutional review board. 9. Birati EY, Rame JE. Post-heart transplant complications. Crit
Care Clin. 2014;30(3):629–37.
Statement of Human and Animal Rights 10. Multani A, Moayedi Y, Puing A, Henricksen E, Garvert
This article does not contain any studies with human or animal DW, Gomez CA, Tremblay-Gravel M, Bunce PE, Luikart H,
subjects. Ross H, Khush KK, et al. Recent trends of infectious com-
plications following heart transplantation. Transplantation.
Statement of Informed Consent 2020;104(10):e284–94.
11. Nellore A, Fishman JA. Donor-derived infections and infec-
There are no human subjects in this article and informed consent is tious risk in xenotransplantation and allotransplantation.
not applicable. Xenotransplantation. 2018;25(4):e12423.
12. Griffith BP, Goerlich CE, Singh AK, Rothblatt M, Lau CL,
Declaration of Conflicting Interests Shah A, Lorber M, Grazioli A, Saharia KK, Hong SN, Joseph
The author(s) declared no potential conflicts of interest with respect SM, et al. Genetically modified porcine-to-human cardiac
to the research, authorship, and/or publication of this article. xenotransplantation. N Engl J Med. 2022;387(1):35–44.
13. Mehta SA, Saharia KK, Nellore A, Blumberg EA, Fishman JA.
Infection and clinical xenotransplantation: guidance from the
Funding
Infectious Disease Community of Practice of the American
The author(s) disclosed receipt of the following financial support Society of Transplantation. Am J Transplant. 2023;23(3):309–15.
for the research, authorship, and/or publication of this article: This 14. Gu W, Zeng N, Zhou L, Ge X, Guo X, Yang H. Genomic
work was supported by grants from the National Natural Science organization and molecular characterization of porcine cyto-
Foundation of China (grant no. 82000227). megalovirus. Virology. 2014;460–461:165–72.
Zhou et al 7

15. Lefkowitz EJ, Dempsey DM, Hendrickson RC, Orton RJ, 33. Denner J. Porcine lymphotropic herpesviruses (PLHVs) and
Siddell SG, Smith DB. Virus taxonomy: the database of the xenotranplantation. Viruses. 2021;13(6):1072.
International Committee on Taxonomy of Viruses (ICTV). 34. Dor FJ, Doucette KE, Mueller NJ, Wilkinson RA, Bajwa JA,
Nucleic Acids Res. 2018;46(D1):D708–17. McMorrow IM, Tseng YL, Kuwaki K, Houser SL, Fishman
16. Liu GH, Li RC, Li J, Huang ZB, Xiao CT, Luo W, Ge M, JA, Cooper DKC, et al. Posttransplant lymphoproliferative
Jiang DL, Yu XL. Seroprevalence of porcine cytomegalovi- disease after allogeneic transplantation of the spleen in minia-
rus and sapovirus infection in pigs in Hunan province, China. ture swine. Transplantation. 2004;78(2):286–91.
Arch Virol. 2012;157(3):521–24. 35. Doucette K, Dor FJ, Wilkinson RA, Martin SI, Huang CA,
17. Liu X, Liao S, Zhu L, Xu Z, Zhou Y. Molecular epidemiology Cooper DK, Sachs DH, Fishman JA. Gene expression of
of porcine Cytomegalovirus (PCMV) in Sichuan Province, porcine lymphotrophic herpesvirus-1 in miniature Swine with
China: 2010-2012. PLoS One. 2013;8(6):e64648. posttransplant lymphoproliferative disorder. Transplantation.
18. Plowright W, Edington N, Watt RG. The behaviour of 2007;83(1):87–90.
porcine cytomegalovirus in commercial pig herds. J Hyg. 36. Hartline CB, Conner RL, James SH, Potter J, Gray E, Estrada
1976;76(1):125–35. J, Tector M, Tector AJ, Prichard MN. Xenotransplantation
19. Gollackner B, Mueller NJ, Houser S, Qawi I, Soizic D, Knosalla panel for the detection of infectious agents in pigs.
C, Buhler L, Dor FJ, Awwad M, Sachs DH, Cooper DKC, et al. Xenotrans­plantation. 2018;25(4):e12427.
Porcine cytomegalovirus and coagulopathy in pig-to-primate 37. Mueller NJ, Kuwaki K, Knosalla C, Dor FJ, Gollackner B,
xenotransplantation. Transplantation. 2003;75(11):1841–47. Wilkinson RA, Arn S, Sachs DH, Cooper DK, Fishman JA.
20. Mueller NJ, Kuwaki K, Dor FJ, Knosalla C, Gollackner Early weaning of piglets fails to exclude porcine lympho-
B, Wilkinson RA, Sachs DH, Cooper DK, Fishman JA. tropic herpesvirus. Xenotransplantation. 2005;12(1):59–2.
Reduction of consumptive coagulopathy using porcine cyto- 38. Tucker AW, McNeilly F, Meehan B, Galbraith D,
megalovirus-free cardiac porcine grafts in pig-to-primate McArdle PD, Allan G, Patience C. Methods for the exclu-
xenotransplantation. Transplantation. 2004;78(10):1449–53. sion of circoviruses and gammaherpesviruses from pigs.
21. Denner J, Längin M, Reichart B, Krüger L, Fiebig U, Mokelke Xenotransplantation. 2003;10(4):343–48.
M, Radan J, Mayr T, Milusev A, Luther F, Sorvillo N, et al. 39. Morozov VA, Plotzki E, Rotem A, Barkai U, Denner J.
Impact of porcine cytomegalovirus on long-term orthotopic Extended microbiological characterization of Göttingen
cardiac xenotransplant survival. Sci Rep. 2020;10(1):17531. minipigs: porcine cytomegalovirus and other viruses.
22. Cohen JI. Herpesvirus latency. J Clin Invest. 2020;130(7): Xenotransplantation. 2016;23(6):490–96.
3361–69. 40. Brema S, Lindner I, Goltz M, Ehlers B. Development of a
23. Fiebig U, Holzer A, Ivanusic D, Plotzki E, Hengel H, Neipel recombinant antigen-based ELISA for the sero-detection of
F, Denner J. Antibody cross-reactivity between porcine cyto- porcine lymphotropic herpesviruses. Xenotransplantation.
megalovirus (PCMV) and human herpesvirus-6 (HHV-6). 2008;15(6):357–64.
Viruses. 2017;9(11):317. 41. Plotzki E, Keller M, Ehlers B, Denner J. Immunological
24. Tesini BL, Epstein LG, Caserta MT. Clinical impact of primary methods for the detection of porcine lymphotropic herpesvi-
infection with roseoloviruses. Curr Opin Virol. 2014;9:91–96. ruses (PLHV). J Virol Methods. 2016;233:72–77.
25. Morozov VA, Morozov AV, Denner J. New PCR diagnostic 42. Niu D, Wei HJ, Lin L, George H, Wang T, Lee IH, Zhao HY,
systems for the detection and quantification of porcine cyto- Wang Y, Kan Y, Shrock E, Lesha E, et al. Inactivation of
megalovirus (PCMV). Arch Virol. 2016;161(5):1159–68. porcine endogenous retrovirus in pigs using CRISPR-Cas9.
26. Mao H, Li J, Liao G, Gao M, Yang G, Bao J. The preven- Science. 2017;357(6357):1303–307.
tion strategies of swine viruses related to xenotransplantation. 43. Denner J, Schuurman HJ. High prevalence of recombinant
Virol J. 2023;20(1):121. porcine endogenous retroviruses (PERV-A/Cs) in minipigs: a
27. Morozov VA, Heinrichs G, Denner J. Effective detection of review on origin and presence. Viruses. 2021;13(9):1869.
porcine cytomegalovirus using non-invasively taken samples 44. Lu TF, Sun B, Yu TY, Wu YJ, Zhou J, Wu SG. Porcine
from piglets. Viruses. 2017;9(1):9. endogenous retroviruses: quantification of the viral copy
28. Plotzki E, Keller M, Ivanusic D, Denner J. A new Western number for the four miniature pig breeds in China. Front
blot assay for the detection of porcine cytomegalovirus Microbiol. 2022;13:840347.
(PCMV). J Immunol Methods. 2016;437:37–42. 45. Denner J. How active are porcine endogenous retroviruses
29. Liu X, Zhu L, Shi X, Xu Z, Mei M, Xu W, Zhou Y, Guo (PERVs)? Viruses. 2016;8(8):215.
W, Wang X. Indirect-blocking ELISA for detecting antibod- 46. Güell M, Niu D, Kan Y, George H, Wang T, Lee IH, Wang G,
ies against glycoprotein B (gB) of porcine cytomegalovirus Church G, Yang L. PERV inactivation is necessary to guar-
(PCMV). J Virol Methods. 2012;186(1–2):30–35. antee absence of pig-to-patient PERVs transmission in xeno-
30. Franzo G, Drigo M, Legnardi M, Grassi L, Menandro ML, transplantation. Xenotransplantation. 2017;24(6). https://doi.
Pasotto D, Cecchinato M, Tucciarone CM. Porcine gam- org/10.1111/xen.12366.
maherpesviruses in Italian commercial swine population: 47. Donahue RE, Kessler SW, Bodine D, McDonagh K, Dunbar C,
frequent but harmless. Pathogens. 2021;10(1):47. Goodman S, Agricola B, Byrne E, Raffeld M, Moen R. Helper
31. Krüger L, Böttger J, Huang CA, Denner J. Absence of por- virus induced T cell lymphoma in nonhuman primates after retro-
cine endogenous retrovirus (PERV) production from pig lym- viral mediated gene transfer. J Exp Med. 1992;176(4):1125–35.
phoma cell lines. Virus Res. 2021;295:198286. 48. Al-Shehabi H, Bannert N. PERV induces CXCL10 in human
32. Chmielewicz B, Goltz M, Franz T, Bauer C, Brema S, monocytes and monocyte-derived primary cells. Intervirology.
Ellerbrok H, Beckmann S, Rziha HJ, Lahrmann KH, Romero 2023;66:30–40.
C, Ehlers B. A novel porcine gammaherpesvirus. Virology. 49. Denner J. Porcine endogenous retroviruses and xenotrans-
2003;308(2):317–29. plantation, 2021. Viruses. 2021;13(11):2156.
8 Cell Transplantation

50. Matsumoto S, Tan P, Baker J, Durbin K, Tomiya M, Azuma reverse transcriptase to nucleoside and non-nucleoside inhibitors.
K, Doi M, Elliott RB. Clinical porcine islet xenotransplan- Cell Mol Life Sci. 2002;59(12):2184–90.
tation under comprehensive regulation. Transplant Proc. 68. Fiebig U, Stephan O, Kurth R, Denner J. Neutralizing anti-
2014;46(6):1992–95. bodies against conserved domains of p15E of porcine endoge-
51. Denner J. Why was PERV not transmitted during preclinical nous retroviruses: basis for a vaccine for xenotransplantation?
and clinical xenotransplantation trials and after inoculation of Virology. 2003;307(2):406–13.
animals? Retrovirology. 2018;15(1):28. 69. Kaulitz D, Fiebig U, Eschricht M, Wurzbacher C, Kurth R,
52. Patience C, Takeuchi Y, Weiss RA. Infection of human Denner J. Generation of neutralising antibodies against porcine
cells by an endogenous retrovirus of pigs. Nat Med. 1997;3(3): endogenous retroviruses (PERVs). Virology. 2011;411(1):
282–86. 78–86.
53. Yang L, Güell M, Niu D, George H, Lesha E, Grishin D, Aach 70. Denner J, Mihica D, Kaulitz D, Schmidt CM. Increased
J, Shrock E, Xu W, Poci J, Cortazio R, et al. Genome-wide titers of neutralizing antibodies after immunization with both
inactivation of porcine endogenous retroviruses (PERVs). envelope proteins of the porcine endogenous retroviruses
Science. 2015;350(6264):1101–104. (PERVs). Virol J. 2012;9:260.
54. Moalic Y, Blanchard Y, Félix H, Jestin A. Porcine endog- 71. Egerer S, Fiebig U, Kessler B, Zakhartchenko V, Kurome M,
enous retrovirus integration sites in the human genome: fea- Reichart B, Kupatt C, Klymiuk N, Wolf E, Denner J, Bähr A.
tures in common with those of murine leukemia virus. J Virol. Early weaning completely eliminates porcine cytomegalovi-
2006;80(22):10980–88. rus from a newly established pig donor facility for xenotrans-
55. Michie C. Xenotransplantation, endogenous pig retroviruses plantation. Xenotransplantation. 2018;25(4):e12449.
and the precautionary principle. Trends Mol Med. 2001;7(2): 72. Geng Y, Zhao C, Guo T, Xu Y, Wang X, Huang W, Liu H,
62–63. Wang Y. Detection of hepatitis E virus in raw pork and pig
56. Jhelum H, Bender M, Reichart B, Mokelke M, Radan J, viscera as food in Hebei province of China. Foodborne Pathog
Neumann E, Krabben L, Abicht JM, Kaufer B, Längin M, Dis. 2019;16(5):325–30.
Denner J. Evidence for microchimerism in baboon recipients 73. Nantel-Fortier N, Letellier A, Lachapelle V, Fravalo P,
of pig hearts. Viruses. 2023;15(7):1618. L’Homme Y, Brassard J. Detection and phylogenetic analy-
57. Denner J. Microchimerism, PERV and xenotransplantation. sis of the hepatitis E virus in a Canadian Swine Production
Viruses. 2023;15(1):190. Network. Food Environ Virol. 2016;8(4):296–304.
58. Chen JQ, Zhang MP, Tong XK, Li JQ, Zhang Z, Huang F, 74. Noordergraaf J, Schucker A, Martin M, Schuurman HJ,
Du HP, Zhou M, Ai HS, Huang LS. Scan of the endoge- Ordway B, Cooley K, Sheffler M, Theis K, Armstrong C,
nous retrovirus sequences across the swine genome and sur- Klein L, Hansen D, et al. Pathogen elimination and prevention
vey of their copy number variation and sequence diversity within a regulated, Designated Pathogen Free, closed pig herd
among various Chinese and Western pig breeds. Zool Res. for long-term breeding and production of xenotransplantation
2022;43(3):423–41. materials. Xenotransplantation. 2018;25(4):e12428.
59. Denner J. Sensitive detection systems for infectious agents in 75. Agrawal N, Dasaradhi PV, Mohmmed A, Malhotra P,
xenotransplantation. Xenotransplantation. 2020;18:e12594. Bhatnagar RK, Mukherjee SK. RNA interference: biol-
60. Stephan O, Schwendemann J, Specke V, Tacke SJ, Boller K, ogy, mechanism, and applications. Microbiol Mol Biol Rev.
Denner J. Porcine endogenous retroviruses (PERVs): genera- 2003;67(4):657–85.
tion of specific antibodies, development of an immunoperoxi- 76. Karlas A, Kurth R, Denner J. Inhibition of porcine endog-
dase assay (IPA) and inhibition by AZT. Xenotransplantation. enous retroviruses by RNA interference: increasing the safety
2001;8(4):310–16. of xenotransplantation. Virology. 2004;325(1):18–23.
61. Denner J, Schuurman HJ, Patience C. The International 77. Dieckhoff B, Petersen B, Kues WA, Kurth R, Niemann H,
Xenotransplantation Association consensus statement on con- Denner J. Knockdown of porcine endogenous retrovirus
ditions for undertaking clinical trials of porcine islet products (PERV) expression by PERV-specific shRNA in transgenic
in type 1 diabetes—chapter 5: strategies to prevent transmis- pigs. Xenotransplantation. 2008;15(1):36–45.
sion of porcine endogenous retroviruses. Xenotransplantation. 78. Ramsoondar J, Vaught T, Ball S, Mendicino M, Monahan J, Jobst
2009;16(4):239–48. P, Vance A, Duncan J, Wells K, Ayares D. Production of trans-
62. Denner J. Virus safety of xenotransplantation. Viruses. genic pigs that express porcine endogenous retrovirus small
2022;14(9):1926. interfering RNAs. Xenotransplantation. 2009;16(3):164–80.
63. Denner J. Xenotransplantation and porcine cytomegalovirus. 79. Semaan M, Kaulitz D, Petersen B, Niemann H, Denner J.
Xenotransplantation. 2015;22(5):329–35. Long-term effects of PERV-specific RNA interference in
64. Fryer JF, Griffiths PD, Emery VC, Clark DA. Susceptibility transgenic pigs. Xenotransplantation. 2012;19(2):112–21.
of porcine cytomegalovirus to antiviral drugs. J Antimicrob 80. Sypniewski D, Bednarek I, Matczyńska D, Gałka S, Loch T,
Chemother. 2004;53(6):975–80. Sołtysik D, Machnik G, Nowak E. Modulation of porcine
65. Argaw T, Colon-Moran W, Wilson C. Susceptibility of endogenous retroviruses (PERVs) expression in vitro by
porcine endogenous retrovirus to anti-retroviral inhibitors. shRNA in the presence of cyclosporine A and dexametha-
Xenotransplantation. 2016;23(2):151–58. sone. Ann Transplant. 2012;17(4):92–107.
66. Denner J. Can antiretroviral drugs be used to treat porcine 81. Fischer K, Kraner-Scheiber S, Petersen B, Rieblinger B,
endogenous retrovirus (PERV) infection after xenotransplan- Buermann A, Flisikowska T, Flisikowski K, Christan S,
tation? Viruses. 2017;9(8):213. Edlinger M, Baars W, Kurome M, et al. Efficient production of
67. Wilhelm M, Fishman JA, Pontikis R, Aubertin AM, Wilhelm FX. multi-modified pigs for xenotransplantation by “combineer-
Susceptibility of recombinant porcine endogenous retrovirus ing,” gene stacking and gene editing. Sci Rep. 2016;6:29081.
Zhou et al 9

82. Semaan M, Ivanusic D, Denner J. Cytotoxic effects during 94. Lee KW, Park SSW, Kim DS, Choi K, Shim J, Kim J, Kim
knock out of multiple porcine endogenous retrovirus (PERV) SJ, Park JB. Auxiliary liver xenotransplantation technique in
sequences in the pig genome by zinc finger nucleases (ZFN). a transgenic pig-to-non-human primate model: a surgical appro­
PLoS One. 2015;10(4):e0122059. ach to prolong survival. Xenotransplantation. 2023:e12814.
83. Yue Y, Xu W, Kan Y, Zhao HY, Zhou Y, Song X, Wu J, Xiong 95. Air Force Medical University News Network. Xijing Hospital
J, Goswami D, Yang M, Lamriben L, et al. Extensive germ- successfully implemented multiple gene-edited pig-rhesus
line genome engineering in pigs. Nat Biomed Eng. 2021;5(2): monkey xenogeneic liver, heart and kidney organ trans-
134–43. plantation. 2020 Jun 30. https://www.fmmu.edu.cn/news/
84. Zheng S, Zhong H, Zhou X, Chen M, Li W, Zi Y, Chi Y, info/1004/168724.htm. (In Chinese).
Wang J, Zheng W, Zou Q, Lai L, et al. Efficient and safe edit- 96. China Daily. Monkeys receive pig organ transplants.
ing of porcine endogenous retrovirus genomes by multiple- 2022 Nov 1. https://www.chinadaily.com.cn/a/202211/01/
site base-editing editor. Cells. 2022;11(24):3975. WS636071a4a310fd2b29e7f962.html.
85. Mohiuddin MM, Singh AK, Corcoran PC, Thomas Iii ML, 97. Wicomb WN, Cooper DK, Lanza RP, Novitzky D, Isaacs
Clark T, Lewis BG, Hoyt RF, Eckhaus M, Pierson Iii RN, S. The effects of brain death and 24 hours’ storage by hypo-
Belli AJ, Wolf E, et al. Chimeric 2C10R4 anti-CD40 antibody thermic perfusion on donor heart function in the pig. J Thorac
therapy is critical for long-term survival of GTKO.hCD46. Cardiovasc Surg. 1986;91(6):896–909.
HTBM pig-to-primate cardiac xenograft. Nat Commun. 98. Zetina-Tun H, Lezama-Urtecho C, Careaga-Reyna G.
2016;7:11138. [Routine hormonal therapy in the heart transplant donor]. Cir
86. Längin M, Mayr T, Reichart B, Michel S, Buchholz S, Guethoff Cir. 2016;84(3):230–34.
S, Dashkevich A, Baehr A, Egerer S, Bauer A, Mihalj M, et 99. Novitzky D, Cooper DK, Morrell D, Isaacs S. Change from
al. Consistent success in life-supporting porcine cardiac xeno- aerobic to anaerobic metabolism after brain death, and rever-
transplantation. Nature. 2018;564(7736):430–33. sal following triiodothyronine therapy. Transplantation. 1988;
87. Reichart B, Längin M, Radan J, Mokelke M, Buttgereit I, 45(1):32–36.
Ying J, Fresch AK, Mayr T, Issl L, Buchholz S, Michel S, 100. Novitzky D, Cooper DK, Reichart B. Hemodynamic and met-
et al. Pig-to-non-human primate heart transplantation: the abolic responses to hormonal therapy in brain-dead potential
final step toward clinical xenotransplantation? J Heart Lung organ donors. Transplantation. 1987;43(6):852–54.
Transplant. 2020;39(8):751–57. 101. Wolbrom DH, Kim JI, Griesemer A. The road to xenotrans-
88. Mohiuddin MM, Goerlich CE, Singh AK, Zhang T, Tatarov plantation. Curr Opin Organ Transplant. 2023;28(2):65–70.
I, Lewis B, Sentz F, Hershfeld A, Braileanu G, Odonkor 102. Reichart B, Längin M, Denner J, Schwinzer R, Cowan PJ,
P, Strauss E, et al. Progressive genetic modifications of Wolf E. Pathways to clinical cardiac xenotransplantation.
porcine cardiac xenografts extend survival to 9 months. Transplantation. 2021;105(9):1930–43.
Xenotransplantation. 2022;29(3):e12744. 103. Meier RPH, Longchamp A, Mohiuddin M, Manuel O, Vrakas
89. NYU Langone Health. Successful heart xenotransplant exper- G, Maluf DG, Buhler LH, Muller YD, Pascual M. Recent
iments at NYU Langone set protocol for pig-to-human organ progress and remaining hurdles toward clinical xenotrans-
transplants. 2022 Jul 9. https://nyulangone.org/news/success- plantation. Xenotransplantation. 2021;28(3):e12681.
ful-heart-xenotransplant-experiments-nyu-langone-set-proto- 104. Carrier AN, Verma A, Mohiuddin M, Pascual M, Muller
col-pig-human-organ-transplants. YD, Longchamp A, Bhati C, Buhler LH, Maluf DG, Meier
90. Kim SC, Mathews DV, Breeden CP, Higginbotham LB, RPH. Xenotransplantation: a new era. Front Immunol. 2022;
Ladowski J, Martens G, Stephenson A, Farris AB, Strobert 13:900594.
EA, Jenkins J, Walters EM, et al. Long-term survival of pig- 105. The University of Alabama at Birmingham. Porcine kidney
to-rhesus macaque renal xenografts is dependent on CD4 T xenotransplantation in patients with end-stage kidney disease;
cell depletion. Am J Transplant. 2019;19(8):2174–85. 2023. https://clinicaltrials.gov/ct2/show/NCT05340426?recrs=a
91. Montgomery RA, Stern JM, Lonze BE, Tatapudi VS, Mangiola bdef&type=Intr&cond=xenotransplantation&draw=10&rank=5.
M, Wu M, Weldon E, Lawson N, Deterville C, Dieter RA, 106. Allcock RJN, Jennison AV, Warrilow D. Towards a uni-
Sullivan B, et al. Results of two cases of pig-to-human kidney versal molecular microbiological test. J Clin Microbiol.
xenotransplantation. N Engl J Med. 2022;386(20):1889–98. 2017;55(11):3175–82.
92. Porrett PM, Orandi BJ, Kumar V, Houp J, Anderson D, Cozette 107. Deurenberg RH, Bathoorn E, Chlebowicz MA, Couto N,
Killian A, Hauptfeld-Dolejsek V, Martin DE, Macedon S, Ferdous M, García-Cobos S, Kooistra-Smid AM, Raangs
Budd N, Stegner KL, et al. First clinical-grade porcine kid- EC, Rosema S, Veloo AC, Zhou K, et al. Application of next
ney xenotransplant using a human decedent model. Am J generation sequencing in clinical microbiology and infection
Transplant. 2022;22(4):1037–53. prevention. J Biotechnol. 2017;243:16–24.
93. Zhang X, Wang Q, Zhao J, Li X, Peng W, Yang Z, Lin Z, 108. Greninger AL. The challenge of diagnostic metagenomics.
Yang L, Ding R, Tao K, Dou K. The resurgent landscape of Expert Rev Mol Diagn. 2018;18(7):605–15.
xenotransplantation of pig organs in nonhuman primates. Sci 109. Fishman JA. Infection in xenotransplantation: opportunities and
China Life Sci. 2021;64(5):697–708. challenges. Curr Opin Organ Transplant. 2019;24(5):527–34.

You might also like