Review Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Transplant. Jun 24, 2014; 4(2): 43-56
Published online Jun 24, 2014. doi: 10.5500/wjt.v4.i2.43
Selecting suitable solid organ transplant donors: Reducing the risk of donor-transmitted infections
Christopher S Kovacs Jr, Christine E Koval, Steven D Mawhorter, Kyle D Brizendine, Eric D Cober, Cyndee Miranda, Rabin K Shrestha, Lucileia Teixeira, Sherif B Mossad, Department of Infectious Diseases, Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, United States
David van Duin, Division of Infectious Diseases, Immunocompromised Host Infectious Disease Program, University of North Carolina, Chapel Hill, NC 27599, United States
Amanda Guedes de Morais, Infectious Disease and Internal Medicine, Augusta Health, Fishersville, VA 22939, United States
Blanca E Gonzalez, Department of Pediatric Infectious Diseases, Cleveland Clinic Children’s, Cleveland, OH 44195, United States
Robin K Avery, Division of Infectious Disease (Transplant/Oncology), Johns Hopkins, Baltimore, MD 21205, United States
Author contributions: Avery RK created the idea of an institutional approach for donor suitability; Koval CE, van Duin D, Mawhorter SD, Brizendine KD, Guedes de Morais A, Gonzalez BE, Cober ED, Miranda C, Shrestha RK, Teixeira L and Mossad S wrote material from which sections were synthesized and reviewed and approved the final draft; Kovacs Jr CS and Mossad SB coordinated and synthesized data, wrote, and compiled the final submitted draft.
Correspondence to: Sherif B Mossad, MD, Department of Infectious Diseases, Medicine Institute, Cleveland Clinic, 9500 Euclid Ave, Mailstop G-21, room 131, Cleveland, OH 44195, United States. mossads@ccf.org
Telephone: +1-216-4452572 Fax: +1-216-4459446
Received: December 20, 2013
Revised: March 21, 2014
Accepted: May 14, 2014
Published online: June 24, 2014

Abstract

Selection of the appropriate donor is essential to a successful allograft recipient outcome for solid organ transplantation. Multiple infectious diseases have been transmitted from the donor to the recipient via transplantation. Donor-transmitted infections cause increased morbidity and mortality to the recipient. In recent years, a series of high-profile transmissions of infections have occurred in organ recipients prompting increased attention on the process of improving the selection of an appropriate donor that balances the shortage of needed allografts with an approach that mitigates the risk of donor-transmitted infection to the recipient. Important advances focused on improving donor screening diagnostics, using previously excluded high-risk donors, and individualizing the selection of allografts to recipients based on their prior infection history are serving to increase the donor pool and improve outcomes after transplant. This article serves to review the relevant literature surrounding this topic and to provide a suggested approach to the selection of an appropriate solid organ transplant donor.

Key Words: Donor selection, Infection, Transplantation, Mass screening, Treatment outcome

Core tip: The literature surrounding preventing donor-transmitted infections in solid organ transplant recipients has increased greatly in the last decade. Increased emphasis has been placed on improved diagnostics for screening of deceased donors. Importance has been placed on using donors who were previously thought to be high risk for transmitting infections to recipients and mitigating the risk to such recipients in an effort to increase the donor pool. Initiating the discussion around using human immunodeficiency virus (HIV) infected donors for HIV infected recipients has important implications for addressing the problem of allograft shortages.



INTRODUCTION

Selection of the appropriate donor is the cornerstone of achieving a positive outcome after solid organ transplantation (SOT). This selection requires screening potential donors for infectious diseases that can be transmitted to the allograft recipient[1]. Screening for transmissible infections allows timely disqualification of a donor if the risk of developing illness in the recipient is deemed prohibitive. Screening also allows risk reduction by identifying and actively treating infection in the donor prior to procurement or preemptively treating the recipient following transplantation. Selecting the suitable donor is of paramount importance to reducing the risk of infectious morbidity and mortality from donor-transmitted infections (DTI).

It has become necessary to consider donors who may have active infection, high-risk infectious serologic profile, or high-risk behavior for human immunodeficiency virus (HIV), hepatitis C virus (HCV), or hepatitis B virus (HBV) infection at the time of donation due to an inadequate supply of needed allografts[2]. As more patients rely on organ transplantation to manage end-stage disease processes, the available donor pool will only shrink further. Important, evidence-based, decisions regarding risk stratification and risk vs benefit analyses are needed in order to increase the donor pool. The risk of death while on the waiting list for many organs needs to be cautiously weighed with the risk for mortality after transplant when considering using expanded donor criteria in order to first do no harm to the recipient (Table 1)[3-8].

Table 1 Mortality figures by type of transplant for 2010 according to the Scientific Registry of Transplant Recipients 2011 Annual Report1.
Organtansplanted2Waiting list mortality incidence density3 (deaths per 1000 patient-years)1 yr posttransplant mortality incidence density (deaths per 1000 patient-years)
Kidney56.534.9
Liver115.6123.7
Intestine71.6193.5
Heart115.891.8
Lung154.1164.2

A number of incidents of DTI have brought this topic to the forefront of attention, as renewed evaluations of the donor screening process have been undertaken. Recent cases of rabies, lymphocytic choriomeningitis virus (LCM), West Nile virus (WNV), HIV, and HCV have all been confirmed as donor transmitted[9-13]. In 2005, the Disease Transmission Advisory Committee (DTAC) was created to aid the Organ Procurement and Transplantation Network/United Network for Organ Sharing in identifying and reviewing potential DTI. This committee has served an essential role in systematizing the collection and evaluation of nationwide information about suspected DTI. This includes: a thorough review of each case by an expert appointed by the committee, facilitation of communication between centers, and tabulating information to a growing database that provides critical information about donor derived risks[14-16]. Extensive deceased donor testing is often not feasible given the time constraints in which such screening must be carried out. Concerns exist regarding sensitivity of tests used for pathogens such as HIV and HCV, which may be negative prior to antibody production[17]. Infections that are reliant on microbiologic methods to diagnose, such as donor blood and urine cultures, may not be resulted until after transplant has taken place. New technologies and donor screening strategies using nucleic acid amplification testing (NAAT) may help provide important information earlier, but developing approaches on how best to utilize these tests has been controversial[1,18,19].

Multiple pathogens have been shown to have the potential to be transmitted by SOT[20,21]. DTIs are estimated to occur in 0.2%-1.7% of all transplant procedures, with varying morbidity and mortality[22,23]. Bacterial, mycobacterial, viral, fungal, and parasitic pathogens all need to be contemplated by the transplant physician when called for opinion regarding donor suitability. This article serves to summarize the current literature about commonly encountered DTI and to offer an approach for decisions regarding donor suitability (Table 2).

Table 2 Approach to selecting suitable donors for solid organ transplantation.
InfectionsDiagnostic toolsTreatment considerations
BacteremiaBlood culturesTreat donor 24 h
AntibiogramTailored recipient therapy in posttransplant period
Resistant bacteriaBlood culturesTailored donor and recipient therapy
Sterile site cultures
Antibiogram
MeningoencephalitisCSF analysisTailored therapy if meningitis only
CSF culture and stain
Cryptococcus antigen
NAAT
SyphilisTreponemal testingTreat recipients as late latent syphilis
Nontreponemal testing
Viral hepatitisSerologic evaluation NAATProphylaxis Tailored therapy HBIG Antivirals
InfluenzaInfluenza testingNeuraminidase inhibitor
Respiratory virus PCR
HTLV 1/2Routine screening not recommendedNo effective treatment, surveillance for recipients of positive donors
Candida infectionBlood cultures Sterile site cultures AntibiogramAntifungal treatment of donor
Treat colonization in certain settings
CryptococcosisCSF cryptococcal antigenAntifungal treatment of donors prior to donation
Serum cryptococcal antigen
Endemic fungiUrine antigen testingAntifungal treatment of donors prior to donation
Serologic evaluation
Sterile site culture
Histologic evaluation
SchistosomiasisStool examinationTreat living donor successfully prior to donation
Serologic evaluation
Rectal biopsy
StrongyloidiasisSerologic evaluationTreat recipients from positive donors
Stool examination
Chagas diseaseEnzyme immunoassayTreat recipient for positive surveillance testing
Radioimmune precipitation assay
BACTERIAL INFECTIONS

Transplantation of allografts taken from donors with underlying sepsis syndrome of unknown etiology is not recommended. Bacterial DTIs have been linked to increased morbidity and mortality as well as allograft loss[24-26]. As previously mentioned, however, underlying bacteremia in the donor may not be recognized until after transplantation has occurred. In one study, 60% of bacteremic donors were afebrile during the 24-h period prior to organ procurement[27]. The outcome of allograft donation from a bacteremic donor depends on the type of bacteria causing infection, previous antimicrobial therapy in the donor prior to organ procurement, and timely recognition of donor bacteremia so therapy can be instituted in the recipient[28,29].

An estimated 5% of organ donors have unrecognized bacteremia at the time of donation[27,30]. Some studies have shown that use of organs from bacteremic donors, especially when the organism is community acquired and not highly resistant to antimicrobials, is not associated with higher incidence of allograft dysfunction[27,30,31]. Thirty-day graft and patient survival for recipients of organs from bacteremic donors were not significantly different than those who received organs from non-bacteremic donors[30]. Recipients included in these series had been given broad-spectrum antibiotics during the perioperative period and were given tailored antibiotic therapy once donor bacteremia was identified. This suggests that allografts from bacteremic donors are suitable for transplantation if the donor is on appropriate antibiotic therapy for ≥ 24 h and if tailored antibiotic therapy can be initiated in the recipient in a timely manner. Recipients should be treated for a minimum of 7 d, depending on the posttransplant course and perhaps longer if the pathogen has the potential to disrupt an anastomosis or seed an endovascular source. In the event a donor is being treated for endocarditis, the recipient should receive organism-specific antimicrobial therapy for at least 2 wk, and if the organism is Staphylococcus aureus, 6 wk of therapy is appropriate[32]. If donor cultures are repeatedly positive for pathogenic bacteria or yeast, then additional consent from the recipient and/or family should be obtained. Surveillance blood cultures of the recipient after transplant are prudent in this situation. Most studies evaluating donor bacteremia excluded donors with sepsis. This may have biased the data by selectively removing pathogens more likely to contribute significantly to posttransplant morbidity and mortality.

An emerging concern is the transmission of multidrug resistant (MDR) bacteria. Management strategies for dealing with these donor-transmitted resistant infections, including methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus (VRE) species, and MDR gram-negative bacteria are not well established[33]. Resistant gram-positive bacteria are frequently encountered in the donor prior to organ procurement. Although less virulent gram-positive bacteria, such as coagulase-negative staphylococci are seemingly less likely to be transmitted from bacteremic donors and are less associated with poorer outcome after transplant, other more virulent gram-positive organisms such as VRE and MRSA do remain a source of concern regarding donor suitability[28]. MRSA colonization of an individual has been shown to increase their risk for infection[34]. Risk factors for MRSA infection and colonization include prolonged hospitalization, exposure to broad-spectrum antibiotics, intensive care unit (ICU) admission, and the presence of a central venous catheter, all of which are often present in deceased organ donors[35]. MRSA colonization in a donor should not prevent acceptance of the allograft; however, perioperative antibiotics should be adjusted to account for the potential increase in recipient infection risk. Mortality from deep-seated MRSA infection associated with bacteremia after transplant has been in excess of 80%[29]. Allografts from donors with deep-seated MRSA infections should only be accepted if the donor has been on appropriate antibiotic therapy for ≥48 h. If the potential allograft is the site of infection, the organ should be rejected. Vancomycin-intermediate Staphylococcus aureus and vancomycin-resistant Staphylococcus aureus infections in the transplant population have not yet been reported[36]. Donor infection with these isolates should exclude them from donation. VRE is another common pathogen, specifically in the setting of transplantation of an intra-abdominal organ. Risk factors for VRE are similar to MRSA, and general guidelines for donor suitability pertaining to MRSA should be applied to reduce recipient risk for VRE infection[37].

Impact of infection with MDR gram-negative bacteria in transplant recipients is of special concern. Literature suggests that survival in transplant recipients with such infections is decreased[38]. These infections are problematic given limited antimicrobial options, need for potentially more toxic antimicrobials, more potential drug interactions, and fewer drugs in the developmental pipeline[33]. Transplant patients are especially vulnerable to infections with these organisms given end-stage disease processes, extensive healthcare contact before and after transplant, and the need for immunosuppression after transplant to maintain graft function. The most common MDR gram-negative infections encountered in the transplant population are carbapenem-resistant Enterobacteriaceae (CRE), carbapenem-resistant Acinetobacter baumannii (CRAB), and Pseudomonas species resistant to at least two different classes of antimicrobials (MDR). Donors with long-term stay in ICU, vasopressor requirement, and prolonged hospitalization are at increased risk for colonization and infection with MDR organisms that can be transmitted to the recipient, even in the absence of overt signs of infection in the donor[39-43]. Studies have shown that using an allograft from a donor with a deep-seated infection from MDR organisms can result in transmission to the recipient even when pathogen directed therapy is used in the recipient[39]. Horizontal transmission within a transplant unit can occur with devastating results. High rates of 30-d mortality have been reported when transplant recipients develop infection with carbapenem-resistant Klebsiella pneumoniae, with infection being a predictor of time-to-death[44,45]. The critical information involves whether the infection is sensitive to a carbapenem. If a donor is colonized with a MDR gram-negative organism that remains sensitive to a carbapenem, he may remain a candidate for donation. A donor with a deep-seated infection involving an organ not being transplanted can be considered only if treated with appropriate antibiotics for ≥ 48 h. Additional consent should be obtained from the recipient and/or family and a plan made to treat the recipient for ≥ 2 wk depending on the clinical course. As a general rule, donor bacteremia with CRE, CRAB, or MDR Pseudomonas infection should eliminate that donor from consideration. Infections stemming from MDR gram-negative organisms no longer susceptible to carbapenems should preclude donation. If a clear case of asymptomatic colonization with a MDR organism is identified in the donor, the allograft may be acceptable, unless noted in the urine or rectal swab of a planned kidney transplant or small bowel transplant, respectively. DTI with these organisms remains an area for study and optimal management strategies for MDR organisms are still to be defined.

Bacterial meningitis and syphilis may be present in a potential organ donor and, as such, may be transmitted to the allograft recipient. The disparity between available allografts and those awaiting transplantation has grown, such that, these two conditions are no longer deemed absolute contraindications for organ donation. Multiple cases of donor-transmitted syphilis have been reported[46-48]. The estimated prevalence of syphilis among potential organ donors based on the incidence in the general population is 0.15%[49]. Transmission of syphilis is a rare event, but if a donor tests positive for this organism additional consent from the recipient and/or family should be obtained. Most experts agree that if the organ is accepted the recipient should be treated with a regimen for late latent syphilis consisting of benzathine penicillin 2.4 million units intramuscularly every week for a total of 3 wk[1]. Syphilis IgG of the recipient should be assessed at time of transplant and at 1, 3, 6, and 12 mo. Patients with documented bacterial meningitis are also no longer considered to be excluded from organ donation, provided that pathogen-directed treatment has been initiated. Several instances of successful allograft procurement have been reported in the literature from donors with microbiologically proven bacterial meningitis[50-54]. Guidelines now recommend accepting an organ if the etiology of the meningitis is Streptococcus pneumoniae, Neisseria meningitidis, Haemophilus influenzae, Escherichia coli, or group B streptococcus. Meningitis must be confirmed as the sole site of infection in the donor and acceptance of donor allografts infected with highly virulent organisms such as Listeria species should be rejected. Ideally, the donor should be receiving appropriate therapy for 48 h prior to procurement with signs of clinical improvement. Additional consent from the recipient and/or family should be obtained and pathogen-directed therapy of the recipient should be continued for at least 2 wk[1].

Cultures of organ procurement fluid (OPF) have been studied as a potential source of DTI. OPF cultures are commonly positive for the growth of bacteria, with low-virulence bacteria such as coagulase-negative staphylococcus and Corynebacterium[55-60]. Studies are variable on whether positive OPF cultures portend an increased risk for posttransplant infection. Cultures of the OPF are rarely available to make donor suitability decisions, but should not prevent organ donation. The exception to this is OPF cultures growing Candida, which may be an important risk factor for graft-transmitted candidiasis[61-64]. The optimal strategy for managing recipients of allografts with positive OPF cultures is not known, but brief tailored treatment of the recipient for growth of virulent organisms is likely indicated[60].

TUBERCULOSIS

Almost 10000 cases of Mycobacterium tuberculosis (TB) infection were reported in the Unites States in 2012. The majority of these cases were in patients who were not born in the United States, but have emigrated from highly endemic areas, highlighting the need for close attention to donor demographics and travel history. It is estimated that rates of tuberculosis in patients from highly endemic areas are 20-74 times the general population with the prevalence of posttransplant tuberculosis approaching 12%[65,66]. Management of tuberculosis in transplant recipients is challenging on many fronts. Diagnosis can be difficult because disease presentation can be atypical, despite ongoing active disease, sputum smears can be negative with low mycobacterial burden, and tuberculin skin testing (TST) and interferon gamma release assays (IGRA) may be falsely negative in the setting of immunosuppression end-stage disease processes[65,67-69]. Treatment is also difficult with concerns for drug toxicity, interactions with immunosuppressive medications, and potential development of drug-resistant tuberculosis. Mycobacterium tuberculosis infection after transplant is associated with 20%-30% mortality rate[67,70].

Most cases of posttransplant tuberculosis are caused by reactivation of latent infection in the recipient following immunosuppressive therapy[71]. Mycobacterium tuberculosis can also be transmitted directly from the allograft to organ recipient[15,72,73]. This fact highlights the necessity of a thoughtful approach to the potential organ donor to limit the risk of a potentially catastrophic posttransplant infectious complication. Table 3 highlights one approach to evaluating the risk of donor-transmitted tuberculosis. There is no firm evidence from randomized clinical trials to make strong recommendations, and each center should factor in the incidence and prevalence of latent TB infection (LTBI) and active TB within their population. Assessment of the donor begins with identifying country of birth, a thorough historical evaluation with emphasis on epidemiological and associated disease-related TB risk factors, prior positive TST/IGRA, review of prior radiographic imaging, and in the case of prior active disease, documentation of completed appropriate anti-tuberculosis treatment. Risk factors for TB in the donor include substance abuse, malnutrition, HIV infection, and close household contact with TB smear-positive individuals[74-77]. Special attention should be paid to donors who have resided in homeless shelters, prisons, or highly endemic areas outside of the United States[78-81]. In donors with low TB risk accompanied by negative radiology, the allograft can be accepted without the need for chemoprophylaxis or additional informed consent on the part of the recipient. Donors, who have had active TB, particularly in the preceding 2 years, have higher relapse potential and increased risk of harboring drug-resistant TB isolates, which, may lead to increased risk of donor-transmitted TB. This should be considered when monitoring and treating recipients of allografts from such donors[69,82].

Table 3 Suggested approach to donor-transmitted Mycobacterium tuberculosis.
Deceased donors
1TB Risk2Suggestive radiology3Donor testing4Donor treatedAccept allograftAdditional consent5Recipient treatmentAdditional recipient testing
LowNoNegativeN/AYesNoneNoneNone
LowYesNegativeNo/YesYesYesChemoprophylaxisNone
LowYesPendingNo/YesNoN/AN/AN/A
ElevatedNoNegativeNo/YesYesYesChemoprophylaxisNone
ElevatedYesNegativeNo/YesYesYesChemoprophylaxisNone
ElevatedYesPendingNo/YesNoN/AN/AN/A
ElevatedYesPositiveNo/YesNoN/AN/AN/A
Prior active TBYesNegativeYesYesYesChemoprophylaxisNone
Prior active TBYesPendingYesYesYesChemoprophylaxisNone
Prior active TBYesPositiveNo/YesNoN/AN/AN/A
Prior active TBYesPositiveNoNoN/AN/AN/A
Active TBYesPositiveNo/YesNoN/AN/AN/A
Living donors
LowNoNegativeN/AYesNoNoneNone
LTBINoPositiveYesNoYesNoneNone
Active TBNoPositiveNoNoN/AN/AN/A
ElevatedYesNegativeNo/YesYesYesChemoprophylaxisNone
VIRAL INFECTIONS

Viral infections are a common cause of morbidity and mortality after SOT. Infections that are potentially donor derived include HIV, HCV, HBV, human T-lymphotropic virus (HTLV- 1 and 2), etiologic agents of viral encephalitis, such WNV, LCM and rabies virus, and viral respiratory pathogens. Cytomegalovirus (CMV) and Epstein-Barr virus (EBV) are commonly donor-transmitted but mainly affect outcomes after the initiation of posttransplant immunosuppression and thus are not addressed in this review. Criteria have been established by the CDC which, when present, may increase the risk of donor transmission of HIV, HBV, and HCV (Table 4)[28]. In the past, many centers have often rejected allografts from such high risk donors. However, availability of improved NAAT testing and closer surveillance monitoring of transplant recipients from CDC-defined high risk donors have allowed these transplants to be undertaken. Aiming to match the allograft to the most appropriate recipient to mitigate the overall risk by improved selection and monitoring has been an overall successful strategy. In such scenarios, additional consent and recipient screening at regular intervals during the first year after transplant should be performed[83].

Table 4 Factors associated with increased risk for human immunodeficiency virus, hepatitis B virus, hepatitis C virus infection and potential donor transmission.
People who have had sex with person known or suspected to have HIV, HBV, or HCV in the preceding 12 mo
MSM in the preceding 12 mo
Women who have had sex with a man with a history of MSM in the preceding 12 mo
People who have had sex in exchange for money or drugs in the preceding 12 mo
People who have had sex with a person who has had sex in exchange for money or drugs in the preceding 12 mo
People who have had sex with a person who has injected drugs for nonmedical reasons in the preceding 12 mo
A child who is ≤ 18 mo of age and born to a mother known to be infected with, or at risk for HIV, HBV or HCV infection
A child who has been breastfed within the preceding 12 mo and the mother is known to be infected with, or at risk for HIV, HBV or HCV infection
People who have injected drugs for nonmedical reasons in the preceding 12 mo
People who have been in lockup, jail, prison or a juvenile correctional facility for ≥ 72 consecutive hours in the preceding 12 h
People who have been newly diagnosed with, or have been treated for, syphilis, gonorrhea, Chlamydia or genital ulcers in the preceding 12 mo
People who have been on hemodialysis in the preceding 12 mo (HCV only)

Viral hepatitis is commonly encountered in both donors and recipients of SOT. HBV infects approximately 400 million people worldwide, with prevalence varying by geographic region[84,85]. As mentioned previously, the ever-enlarging pool of patients awaiting lifesaving transplants has necessitated relaxation of exclusion criteria used to select suitable organ donors. This has led to the usage of allografts taken from donors who have previously had HBV infection (anti-HB core antibody positive donors). The development of de novo hepatitis B infection in recipients of allografts from anti-HBc positive donors has been noted since 1992, but after initially excluding these donors, it has been found that allografts from these donors can be safely used[86-89]. Careful selection of the donor is essential when considering recipients coinfected with HBV and HDV as recurrence of disease is common in this setting and specific posttransplant treatments may need to be implemented to optimize outcomes[90]. HCV is a cause of chronic hepatitis in 3-4 million people in the United States and is the leading indication for liver transplantation[91]. As both HBV and HCV can be transmitted via organ donation, a thorough approach is needed for successful management of the recipient, and an emphasis on aggressive immunization and risk mitigation of transplant candidates prior to transplant should be pursued.

Decisions regarding donor suitability depend on whether living-related partial liver donation is planned and disease status of the donor and recipient at the time of allograft procurement. More stringent, evidence-based guidelines regarding the use of anti-HBc antibody donors are forthcoming, but currently it is felt that allografts from HBV infected donors should preferentially be given to recipients who are hepatitis B surface antigen positive, core antibody positive, or surface antibody positive[92]. In both hepatic and non-hepatic donors, an allograft from a donor with acute hepatitis B infection should not be accepted, regardless of the serologic status of the recipient. Hepatitis B surface antigen positive donors can donate to HB surface antigen positive recipients, but hepatitis B immunoglobulin (HBIG) and antiviral therapy should be given with advanced planning. Donors who are anti-HBc antibody positive and HBsAg negative are acceptable, but additional consent should be obtained from the recipient prior to transplant[65,93-95]. Antiviral treatment should be given at the time of transplant to recipients of liver allografts from donors with prior evidence of HBV infection. HBIG should be administered to liver allograft recipients who lack surface antibody to HBV[96-102]. Non-immune non-hepatic allograft recipients should also receive antiviral prophylaxis if the donor is anti-HBc positive and HBV DNA is detected. HCV infected donors should be precluded from donating an allograft to a HCV naïve recipient[103]. HCV infected hepatic and non-hepatic allografts can be donated to HCV infected recipients with the caveat that donors with HCV genotype 1 infection should preferentially be used for recipients with HCV genotype 1 infection if that donor information is known prior to donation[92,104-108].

Influenza and other respiratory viruses are another potential cause of DTI. Influenza, respiratory syncytial virus (RSV), parainfluenza virus, human metapneumovirus (hMPV), adenovirus, and coronavirus are usually self-limited illnesses in healthy adults but have the potential for significant morbidity and mortality in the SOT population. These viruses cause a wide range of disease, and transplant recipients often have atypical presentations and more severe symptoms[109]. The burden of illness of these viruses follows a seasonal pattern, mainly occurring during the fall and winter months[110]. DTI with these respiratory viruses can increase the risk of secondary bacterial or fungal pneumonia in the recipient, lead to a prolonged period of viral shedding, and potentially contribute to increased risk of allograft rejection in lung transplant recipients[109,111-114]. DTI of respiratory viruses is further complicated by limited treatment options. Influenza and adenovirus have both been reported as DTI with devastating consequences[115-117]. As such, high index of suspicion is needed when evaluating a donor, especially during the peak seasons of respiratory viral infections within the community. During peak seasonal epidemic activity or in the setting of an ongoing pandemic, donors and recipients should be screened for clinical symptoms of an influenza-like illness. Lung and intestinal potential donors who have been diagnosed with influenza within the previous two weeks should be disqualified from donation. Other types of allografts can be accepted if additional consent is obtained, the donor has received anti- influenza treatment, and the recipient is given neuraminidase inhibitor chemoprophylaxis after transplant. Donors of any allograft with influenza diagnosed greater than 2 wk prior to donation, who are adequately treated and no longer symptomatic can be utilized. Oseltamivir resistant influenza diagnosed in any donor should preclude his/her use as a donor[118]. Lung allografts from donors infected with other respiratory viruses should be rejected with the exception of resolved RSV infection with no residual symptoms. Non-lung allografts infected with respiratory viruses other than influenza can be accepted. If lower respiratory tract sampling shows viral respiratory infection other than influenza or radiograph show an infiltrate and that lung allograft is accepted for use in a dire situation, oral ribavirin can be considered as chemoprophylaxis for the recipient[109]. All allografts from donors with adenovirus infection should be rejected as adenovirus infections in the recipient tend to recur in the transplanted organ[117,119].

Additional viral infections that are potentially donor transmitted include HTLV-1/2 and the etiologic agents of viral encephalitis. Although no longer required as a screening test in deceased donors, concerns remain regarding donor-transmission of HTLV-1/2[120,121]. Rapid progression from infection to disease has been noted in transplant recipients, with the development of myelopathic spastic paraparesis and adult T-cell leukemia/lymphoma[122]. Donors who test positive for these viruses should be precluded from allograft donation unless required for an emergent life-threatening situation. If allograft is accepted, additional consent should be obtained, and the recipient should have virus-specific serology and polymerase chain reaction (PCR) testing at the time of transplant and 1, 3, and 12 mo[123]. Allografts from patients with suspected viral encephalitis should not be accepted given the risk of transmission of WNV, rabies, LCM and herpes simplex virus infections[124-126]. This recommendation may also extend to cerebrospinal fluid pleocytosis where bacterial meningitis has not been proven by either culture or antigen testing indicating a specific bacterial pathogen as the cause of infection.

FUNGAL INFECTIONS

Fungal infections often affect the critically ill potential organ donor and, as such, have the potential to be donor-transmitted. Recipient DTIs with Candida species, cryptococcosis, endemic fungal infections, aspergillosis, and non-Aspergillus mold infections have all been documented and, when they occur, are important causes of recipient morbidity and mortality[127].

Outcomes of fungal DTI depend on the type of fungal infection identified, the specific allograft donated, and antifungal susceptibilities of recovered isolates. Infections associated with Candida species may occur in the setting of positive preservation fluid cultures, possibly due to contamination at the time of organ procurement[61,63,128]. Bowel perforation in the donor is another common source of Candida contamination of the allograft[61]. In general, patients with untreated invasive fungal infections should not be used as organ donors. Aspergillus and other invasive mold infections result in significant morbidity and mortality from graft site abscesses and anastomotic infections, despite treatment of both donor and recipient[127]. Renal allografts from donors with candiduria and lung allografts from donors with bronchial cultures positive for Candida species can be used with appropriate treatment. Recipients of lung allograft from a donor with documented Candida colonization of the airways have been shown to benefit from universal prophylaxis with an echinocandin for the prevention of early posttransplant infections; including empyema[127,129]. Treatment of renal allograft recipients from donors with candiduria should consist of a tailored antifungal agent for urinary tract involvement. Urinary levels of fluconazole exceed minimum inhibitory concentration values for most Candida species and can be used in most cases. Therapy should be continued for up to 6 wk depending on whether there is vascular involvement of the urinary tract[62,127,130]. After lung transplant, treatment should be continued until bronchoscopic evaluation confirms the integrity of the bronchial anastomosis[127].

Cryptococcosis can occur in up to 5% of SOT recipients[131]. Most infections after transplant represent reactivation of recipient latent infection, but DTIs do occur in a subset of patients[132,133]. The potential for cryptococcal DTI should be considered when a donor presents with undiagnosed neurological illness, unrecognized meningoencephalitis, or pulmonary nodules in the setting of risk factors for cryptococcosis, such as prior hematologic malignancy, steroid treatment, sarcoidosis, or other cell-mediated immune dysfunction[134]. Cerebrospinal fluid cryptococcal antigen and serum cryptococcal antigen should be obtained from donors who meet these clinical risk factors. Donors with active cryptococcal disease should be excluded from donation. Recovery of Cryptococcus in the recipient should not be treated as contamination or colonization, but should prompt initiation of therapeutic antifungal treatment[135].

Endemic fungal infection should be considered as a potential DTI when donors reside in endemic areas or travel frequently to areas with high incidence of histoplasmosis, blastomycosis, or coccidioidomycosis. These areas include the Ohio and Mississippi river valleys, the Great Lakes region, and Southwestern United States, respectively. Since histoplasmosis occurs in only 0.5% of SOT recipients residing in endemic regions, routine laboratory screening of all donors is not warranted[136]. Donors should be evaluated for a prior history or signs and symptoms compatible with active histoplasmosis. If current concerns or prior history exist, an assessment consisting of agar gel immunodiffusion, complement fixation antibody titers, and urine Histoplasma antigen should be undertaken. The presence of antigenuria, H precipitin bands, or complement fixation antibody titers ≥ 1:32 should lead to rejection of the donor allograft. Coccidioidomycosis is a dimorphic fungus that is endemic in the Southwestern United States, Mexico, Central and South America. Approximately 150000 infections occur annually in the US, with an estimated 1.4%-6.9% of transplant recipients becoming infected[137]. Reactivation of latent infection is the most common mode of posttransplant infection, but multiple cases of DTI have been documented in patients from both endemic and non-endemic areas[138,139]. Patients with active coccidioidomycosis should not be permitted to donate an organ for transplantation. In donors with prior history of coccidioidomycosis, an evaluation should be undertaken to document clearance of infection; including history documenting the resolution of symptoms, resolution of radiographic abnormalities, and at least a 4-fold decrease in antibody titer[140]. Fluconazole or itraconazole can be used for the prevention of DTI in the event that a recipient receives an organ from a donor who in retrospect had evidence of remote infection[141]. Lifelong prophylaxis is indicated following treatment doses for at least one year. Fluconazole at an average daily dose of 200-400 mg can be used depending on whether prophylaxis is primary or secondary[137].

PARASITIC INFECTIONS

With increase in international travel and immigration, potential organ donors have greater risk for parasitic infections not endemic to the United States. Transmission of Chagas disease, schistosomiasis, and Strongyloides has been reported[142-144].

The optimal screening procedure for schistosomiasis in donors from endemic areas has not yet been established. Screening of living donors from endemic areas with fecal parasitological analysis paired with blood Schistosoma antibody detection assay is a reasonable starting point. This can be followed with a stepwise approach including rectal biopsy, liver biopsy, or both depending on the results of the initial screening tests. If stool analysis shows Schistosoma eggs, liver biopsy should be performed regardless of the result of Schistosoma serology. In the situation where Schistosoma eggs are not detected in the stools but the donor is noted to be seropositive for Schistosoma, further investigation with a rectal biopsy is indicated. If rectal biopsy demonstrates Schistosoma eggs, all allografts from this donor should be rejected. If eggs are found on initial screening, living donor treatment with praziquantel should be initiated followed by repeat testing of stools for Schistosoma eggs. Only if repeat stool testing is negative, should the patient be accepted to donate[145].

Screening of both donors and recipients for strongyloidiasis in the pretransplant period is recommended for those at epidemiologic risk and should include both serology and stool studies[146]. A donor with documented strongyloidiasis should not be precluded from donation, but additional consent from the recipient should be obtained. Recipients of organs from such donor should be prophylactically treated with ivermectin.

Chagas disease is an infection caused by the parasite Trypanosoma cruzi. It is endemic to Mexico, Central, and South America but has the potential to cause DTI in the setting of transplantation from a donor from an endemic region to a recipient in a non-endemic country[147]. Most posttransplant infections occurring in recipients from endemic regions occur due to reactivation of latent infection as a result of iatrogenic immunosuppression. Transmission rates from seropositive donors to seronegative recipients are approximately 20% for kidney transplant recipients and 30% for liver transplant recipients. Screening for Chagas disease should be performed on donors who were born or spent significant time living in an endemic country[148]. Donors who have a history of treated Chagas disease should also be screened using the Ortho enzyme immunoassay test (Ortho-Clinical Diagnostics, Inc.; Raritan, New Jersey) and the Abbot Prism Chagas test (Abbott Laboratories; Abbott Park, Illinois). If the initial screening of a living donor is positive, a second confirmatory test should be sent to the CDC; using the radioimmune precipitation assay. Deceased donor testing should also be performed but this information may not be available at the time of transplantation[149]. No allograft should be accepted from a donor who died from acute Chagas disease. When a donor has positive serology for Chagas disease or has a history of treated Chagas disease, organs other than the heart or intestine may be suitable for transplantation with additional consent and posttransplant screening of the recipient. Testing should include T. cruzi PCR and microscopy of blood peripheral smears at predetermined time intervals, or in the event of fever, and when rejection is present. Treatment is only indicated if surveillance testing of the recipient is consistent with T. cruzi infection. Heart or intestinal transplantation from a donor with a positive history or serology for T. cruzi is thought to represent too high of a potential risk for DTI to be acceptable[146,150-152].

CONCLUSION

The demand for allografts for the treatment of end-stage disease processes continues to grow. The need for a thoughtful and thorough approach to donor selection has never been more important in balancing unnecessarily discarding potentially lifesaving organs with reducing infectious complications for the recipient after transplant. Decisions regarding donor acceptability should be made in conjunction with a clinician who has special training and experience in dealing with infections related to transplantation. Donor history and physical examination should be meticulous with an emphasis on documenting current or latent infections that can be transmitted to the recipient. Screening using molecular and microbiological testing should be attempted, as time permits, prior to organ procurement in order to allow for rejection of an unacceptable allograft, or to allow for monitoring and treatment in the recipient. As the need for organs continues to rise, special attention will be focused on ways to expand the donor pool.

Multiple HIV-infected patients die each year awaiting organs that could be provided from living or deceased HIV-infected donors. Approximately 500 HIV positive deceased donors are not currently being utilized to donate organs to HIV-positive recipients[153]. Improvements in antiretroviral therapy and report of successful kidney transplantation from a donor with HIV infection in South Africa make this an interesting, albeit complicated, area for future evaluation and research. A key advancement has recently occurred with the passage of the HIV Organ Policy Equity Act (HOPE Act) on 11/21/2013.

Improved development of NAAT in conjunction with defined and validated algorithms of application may allow for faster and more accurate testing of donor specimens enabling previously excluded donors to be accepted for donation. Focused efforts to reassess the risk of using high-risk donors should be undertaken, and methods for decreasing recipient risk of DTI are imperative. Finally, it is important to continue to build on the substantial contributions to quality and safety made by the DTAC in recent years. Providers should be strongly encouraged to report any possible donor-transmitted event in real time. Critical infrastructure is now in place to investigate potential DTI, to take appropriate action in the treatment of potential recipients at risk, and to analyze indispensable data in the pursuit of evidence-based decision making essential to improving outcomes in this unique patient population.

Footnotes

P- Reviewers: Jaing TH, Ravaioli M S- Editor: Song XX L- Editor: A E- Editor: Wu HL

References
1.  Fischer SA, Lu K. Screening of donor and recipient in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:9-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 87]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
2.  Nadig SN, Bratton CF, Karp SJ. Marginal donors in liver transplantation: expanding the donor pool. J Surg Educ. 2007;64:46-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
3.  Matas AJ, Smith JM, Skeans MA, Lamb KE, Gustafson SK, Samana CJ, Stewart DE, Snyder JJ, Israni AK, Kasiske BL. OPTN/SRTR 2011 Annual Data Report: kidney. Am J Transplant. 2013;13 Suppl 1:11-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 189]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
4.  Kandaswamy R, Stock PG, Skeans MA, Gustafson SK, Sleeman EF, Wainright JL, Carrico RJ, Ghimire V, Snyder JJ, Israni AK. OPTN/SRTR 2011 Annual Data Report: pancreas. Am J Transplant. 2013;13 Suppl 1:47-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 67]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
5.  Kim WR, Stock PG, Smith JM, Heimbach JK, Skeans MA, Edwards EB, Harper AM, Snyder JJ, Israni AK, Kasiske BL. OPTN/SRTR 2011 Annual Data Report: liver. Am J Transplant. 2013;13 Suppl 1:73-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 137]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
6.  Smith JM, Skeans MA, Thompson B, Horslen SP, Edwards EB, Harper AM, Snyder JJ, Israni AK, Kasiske BL. OPTN/SRTR 2011 Annual Data Report: intestine. Am J Transplant. 2013;13 Suppl 1:103-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
7.  Colvin-Adams M, Smith JM, Heubner BM, Skeans MA, Edwards LB, Waller C, Snyder JJ, Israni AK, Kasiske BL. OPTN/SRTR 2011 Annual Data Report: heart. Am J Transplant. 2013;13 Suppl 1:119-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
8.  Valapour M, Paulson K, Smith JM, Hertz MI, Skeans MA, Heubner BM, Edwards LB, Snyder JJ, Israni AK, Kasiske BL. OPTN/SRTR 2011 Annual Data Report: lung. Am J Transplant. 2013;13 Suppl 1:149-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 84]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
9.  Srinivasan A, Burton EC, Kuehnert MJ, Rupprecht C, Sutker WL, Ksiazek TG, Paddock CD, Guarner J, Shieh WJ, Goldsmith C. Transmission of rabies virus from an organ donor to four transplant recipients. N Engl J Med. 2005;352:1103-1111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 350]  [Cited by in F6Publishing: 366]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
10.  Fischer SA, Graham MB, Kuehnert MJ, Kotton CN, Srinivasan A, Marty FM, Comer JA, Guarner J, Paddock CD, DeMeo DL. Transmission of lymphocytic choriomeningitis virus by organ transplantation. N Engl J Med. 2006;354:2235-2249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 424]  [Cited by in F6Publishing: 347]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
11.  Iwamoto M, Jernigan DB, Guasch A, Trepka MJ, Blackmore CG, Hellinger WC, Pham SM, Zaki S, Lanciotti RS, Lance-Parker SE. Transmission of West Nile virus from an organ donor to four transplant recipients. N Engl J Med. 2003;348:2196-2203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 520]  [Cited by in F6Publishing: 436]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
12.  Ison MG, Llata E, Conover CS, Friedewald JJ, Gerber SI, Grigoryan A, Heneine W, Millis JM, Simon DM, Teo CG. Transmission of human immunodeficiency virus and hepatitis C virus from an organ donor to four transplant recipients. Am J Transplant. 2011;11:1218-1225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 111]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
13.  Ahn J, Cohen SM. Transmission of human immunodeficiency virus and hepatitis C virus through liver transplantation. Liver Transpl. 2008;14:1603-1608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 59]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
14.  Ison MG, Nalesnik MA. An update on donor-derived disease transmission in organ transplantation. Am J Transplant. 2011;11:1123-1130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 179]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
15.  Ison MG, Hager J, Blumberg E, Burdick J, Carney K, Cutler J, Dimaio JM, Hasz R, Kuehnert MJ, Ortiz-Rios E. Donor-derived disease transmission events in the United States: data reviewed by the OPTN/UNOS Disease Transmission Advisory Committee. Am J Transplant. 2009;9:1929-1935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 186]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
16.  Nalesnik MA, Woodle ES, Dimaio JM, Vasudev B, Teperman LW, Covington S, Taranto S, Gockerman JP, Shapiro R, Sharma V. Donor-transmitted malignancies in organ transplantation: assessment of clinical risk. Am J Transplant. 2011;11:1140-1147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 148]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
17.  Fishman JA, Strong DM, Kuehnert MJ. Organ and tissue safety workshop 2007: advances and challenges. Cell Tissue Bank. 2009;10:271-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
18.  Humar A, Morris M, Blumberg E, Freeman R, Preiksaitis J, Kiberd B, Schweitzer E, Ganz S, Caliendo A, Orlowski JP. Nucleic acid testing (NAT) of organ donors: is the ‘best’ test the right test? A consensus conference report. Am J Transplant. 2010;10:889-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 142]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
19.  Shafer TJ, Schkade D, Schkade L, Geier SS, Orlowski JP, Klintmalm G. Zero risk tolerance costs lives: loss of transplantable organs due to human immunodeficiency virus nucleic acid testing of potential donors. Prog Transplant. 2011;21:236-247; quiz 248.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Avery RK, Ljungman P. Prophylactic measures in the solid-organ recipient before transplantation. Clin Infect Dis. 2001;33 Suppl 1:S15-S21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 70]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
21.  Fishman JA, Issa NC. Infection in organ transplantation: risk factors and evolving patterns of infection. Infect Dis Clin North Am. 2010;24:273-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 97]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
22.  Grossi PA, Costa AN, Fehily D, Blumberg EA, Kuehnert MJ, Fishman JA, Ison MG, Lattes R, Kotton CN, Lilleri D. Infections and organ transplantation: new challenges for prevention and treatment--a colloquium. Transplantation. 2012;93:S4-S39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
23.  Len O, Gavaldà J, Blanes M, Montejo M, San Juan R, Moreno A, Carratalà J, de la Torre-Cisneros J, Bou G, Cordero E. Donor infection and transmission to the recipient of a solid allograft. Am J Transplant. 2008;8:2420-2425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 57]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
24.  Weber TR, Freier DT, Turcotte JG. Transplantation of infected kidneys: clinical and experimental results. Transplantation. 1979;27:63-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
25.  Berggren H, Berglin E, Kjellman U, Mantovani V, Nilsson B. Successful outcome after massive bleeding in a heart transplant recipient with mycotic aortitis. Case report. Scand J Thorac Cardiovasc Surg. 1994;28:45-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
26.  Nery JR, Weppler D, Ketchum P, Olson L, Fragulidis GP, Khan MF, Webb MG, Miller J, Tzakis AG. Donor infection and primary nonfunction in liver transplantation. Transplant Proc. 1997;29:481-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
27.  Lumbreras C, Sanz F, González A, Pérez G, Ramos MJ, Aguado JM, Lizasoain M, Andrés A, Moreno E, Gómez MA. Clinical significance of donor-unrecognized bacteremia in the outcome of solid-organ transplant recipients. Clin Infect Dis. 2001;33:722-726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 100]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
28.  Ison MG, Grossi P. Donor-derived infections in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:22-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 116]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
29.  Singh N. Impact of donor bacteremia on outcome in organ transplant recipients. Liver Transpl. 2002;8:975-976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
30.  Freeman RB, Giatras I, Falagas ME, Supran S, O’Connor K, Bradley J, Snydman DR, Delmonico FL. Outcome of transplantation of organs procured from bacteremic donors. Transplantation. 1999;68:1107-1111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 145]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
31.  Kubak BM, Gregson AL, Pegues DA, Leibowitz MR, Carlson M, Marelli D, Patel J, Laks H, Kobashigawa JA. Use of hearts transplanted from donors with severe sepsis and infectious deaths. J Heart Lung Transplant. 2009;28:260-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
32.  Caballero F, Lopez-Navidad A, Perea M, Cabrer C, Guirado L, Solà R. Successful liver and kidney transplantation from cadaveric donors with left-sided bacterial endocarditis. Am J Transplant. 2005;5:781-787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 26]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
33.  Sifri CD, Ison MG. Highly resistant bacteria and donor-derived infections: treading in uncharted territory. Transpl Infect Dis. 2012;14:223-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
34.  Wertheim HF, Melles DC, Vos MC, van Leeuwen W, van Belkum A, Verbrugh HA, Nouwen JL. The role of nasal carriage in Staphylococcus aureus infections. Lancet Infect Dis. 2005;5:751-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1591]  [Cited by in F6Publishing: 1633]  [Article Influence: 85.9]  [Reference Citation Analysis (0)]
35.  Asensio A, Guerrero A, Quereda C, Lizán M, Martinez-Ferrer M. Colonization and infection with methicillin-resistant Staphylococcus aureus: associated factors and eradication. Infect Control Hosp Epidemiol. 1996;17:20-28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 80]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
36.  Garzoni C, Vergidis P. Methicillin-resistant, vancomycin-intermediate and vancomycin-resistant Staphylococcus aureus infections in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:50-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
37.  Patel G, Snydman DR. Vancomycin-resistant Enterococcus infections in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:59-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
38.  van Duin D, van Delden C. Multidrug-resistant gram-negative bacteria infections in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:31-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 89]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
39.  Ariza-Heredia EJ, Patel R, Blumberg EA, Walker RC, Lewis R, Evans J, Sankar A, Willliams MD, Rogers J, Milano C. Outcomes of transplantation using organs from a donor infected with Klebsiella pneumoniae carbapenemase (KPC)-producing K. pneumoniae. Transpl Infect Dis. 2012;14:229-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
40.  Watkins AC, Vedula GV, Horan J, Dellicarpini K, Pak SW, Daly T, Samstein B, Kato T, Emond JC, Guarrera JV. The deceased organ donor with an “open abdomen”: proceed with caution. Transpl Infect Dis. 2012;14:311-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
41.  Goldberg E, Bishara J, Lev S, Singer P, Cohen J. Organ transplantation from a donor colonized with a multidrug-resistant organism: a case report. Transpl Infect Dis. 2012;14:296-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 47]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
42.  Simkins J, Muggia V. Favorable outcome in a renal transplant recipient with donor-derived infection due to multidrug-resistant Pseudomonas aeruginosa. Transpl Infect Dis. 2012;14:292-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
43.  Martins N, Martins IS, de Freitas WV, de Matos JA, Magalhães AC, Girão VB, Dias RC, de Souza TC, Pellegrino FL, Costa LD. Severe infection in a lung transplant recipient caused by donor-transmitted carbapenem-resistant Acinetobacter baumannii. Transpl Infect Dis. 2012;14:316-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 49]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
44.  Bergamasco MD, Barroso Barbosa M, de Oliveira Garcia D, Cipullo R, Moreira JC, Baia C, Barbosa V, Abboud CS. Infection with Klebsiella pneumoniae carbapenemase (KPC)-producing K. pneumoniae in solid organ transplantation. Transpl Infect Dis. 2012;14:198-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 119]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
45.  Kalpoe JS, Sonnenberg E, Factor SH, del Rio Martin J, Schiano T, Patel G, Huprikar S. Mortality associated with carbapenem-resistant Klebsiella pneumoniae infections in liver transplant recipients. Liver Transpl. 2012;18:468-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 111]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
46.  Cortes NJ, Afzali B, MacLean D, Goldsmith DJ, O’Sullivan H, Bingham J, Lewis DA, MacMahon E, Tong CY, Koffman G. Transmission of syphilis by solid organ transplantation. Am J Transplant. 2006;6:2497-2499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
47.  Marek A, Inkster T. A syphilis-positive organ donor -- management of the cardiac transplant recipient: a case report and review of the literature. Sex Transm Dis. 2012;39:485-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
48.  Tariciotti L, Das I, Dori L, Perera MT, Bramhall SR. Asymptomatic transmission of Treponema pallidum (syphilis) through deceased donor liver transplantation. Transpl Infect Dis. 2012;14:321-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
49.  Gibel LJ, Sterling W, Hoy W, Harford A. Is serological evidence of infection with syphilis a contraindication to kidney donation? Case report and review of the literature. J Urol. 1987;138:1226-1227.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Caballero F, Puig M, Santos JA, Deulofeu R, Ballarín J, Charco R, Leal J. Successful transplantation of organs from a donor with postneurosurgical meningitis caused by Escherichia coli. Transplantation. 2012;93:e11-e13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
51.  Cantarovich M, Tchervenkov J, Loertscher R. Transplantation of kidneys from a donor with Neisseria meningitidis infection. Am J Nephrol. 1993;13:171-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
52.  López-Navidad A, Domingo P, Caballero F, González C, Santiago C. Successful transplantation of organs retrieved from donors with bacterial meningitis. Transplantation. 1997;64:365-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 71]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
53.  Issa NC, Patel R. Potential for expansion of the donor pool using liver allografts from donors with bacterial meningitis. Liver Transpl. 2002;8:977-979.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
54.  Bahrami T, Vohra HA, Shaikhrezai K, Tadjkarimi S, Banner N, Amrani M, Yacoub M, Khaghani A. Intrathoracic organ transplantation from donors with meningitis: a single-center 20-year experience. Ann Thorac Surg. 2008;86:1554-1556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
55.  Yansouni CP, Dendukuri N, Liu G, Fernandez M, Frenette C, Paraskevas S, Sheppard DC. Positive cultures of organ preservation fluid predict postoperative infections in solid organ transplantation recipients. Infect Control Hosp Epidemiol. 2012;33:672-680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
56.  Grąt M, Ligocka J, Lewandowski Z, Barski K, Hołówko W, Skalski M, Kornasiewicz O, Usarek P, Zieniewicz K, Młynarczyk G. Incidence, pattern and clinical relevance of microbial contamination of preservation fluid in liver transplantation. Ann Transplant. 2012;17:20-28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
57.  Sauget M, Verdy S, Slekovec C, Bertrand X, Talon D. Bacterial contamination of organ graft preservation solution and infection after transplantation. Transpl Infect Dis. 2011;13:331-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
58.  Ruiz P, Gastaca M, Gonzalez J, Hernandez MJ, Ventoso A, Valdivieso A, Montejo M, Ortiz de Urbina J. Incidence and clinical relevance of bacterial contamination in preservation solution for liver transplantation. Transplant Proc. 2009;41:2169-2171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
59.  Janny S, Bert F, Dondero F, Durand F, Guerrini P, Merckx P, Nicolas-Chanoine MH, Belghiti J, Mantz J, Paugam-Burtz C. Microbiological findings of culture-positive preservation fluid in liver transplantation. Transpl Infect Dis. 2011;13:9-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
60.  Mossad SB, Avery RK, Goormastic M, Hobbs RE, Stewart RW. Significance of positive cultures from donor left atrium and postpreservation fluid in heart transplantation. Transplantation. 1997;64:1209-1210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 15]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
61.  Albano L, Bretagne S, Mamzer-Bruneel MF, Kacso I, Desnos-Ollivier M, Guerrini P, Le Luong T, Cassuto E, Dromer F, Lortholary O. Evidence that graft-site candidiasis after kidney transplantation is acquired during organ recovery: a multicenter study in France. Clin Infect Dis. 2009;48:194-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 87]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
62.  Mai H, Champion L, Ouali N, Hertig A, Peraldi MN, Glotz D, Rondeau E, Costa MA, Snanoudj R, Benoit G. Candida albicans arteritis transmitted by conservative liquid after renal transplantation: a report of four cases and review of the literature. Transplantation. 2006;82:1163-1167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 54]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
63.  Matignon M, Botterel F, Audard V, Dunogue B, Dahan K, Lang P, Bretagne S, Grimbert P. Outcome of renal transplantation in eight patients with Candida sp. contamination of preservation fluid. Am J Transplant. 2008;8:697-700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 55]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
64.  Veroux M, Corona D, Scriffignano V, Caglià P, Gagliano M, Giuffrida G, Gona F, Sciacca A, Giaquinta A, Oliveri S. Contamination of preservation fluid in kidney transplantation: single-center analysis. Transplant Proc. 2010;42:1043-1045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
65.  Muñoz SJ. Use of hepatitis B core antibody-positive donors for liver transplantation. Liver Transpl. 2002;8:S82-S87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
66.  Sakhuja V, Jha V, Varma PP, Joshi K, Chugh KS. The high incidence of tuberculosis among renal transplant recipients in India. Transplantation. 1996;61:211-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 138]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
67.  Singh N, Paterson DL. Mycobacterium tuberculosis infection in solid-organ transplant recipients: impact and implications for management. Clin Infect Dis. 1998;27:1266-1277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 439]  [Cited by in F6Publishing: 455]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
68.  Currie AC, Knight SR, Morris PJ. Tuberculosis in renal transplant recipients: the evidence for prophylaxis. Transplantation. 2010;90:695-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 72]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
69.  Horne DJ, Narita M, Spitters CL, Parimi S, Dodson S, Limaye AP. Challenging issues in tuberculosis in solid organ transplantation. Clin Infect Dis. 2013;57:1473-1482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
70.  Holty JE, Sista RR. Mycobacterium tuberculosis infection in transplant recipients: early diagnosis and treatment of resistant tuberculosis. Curr Opin Organ Transplant. 2009;14:613-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
71.  Subramanian AK, Morris MI. Mycobacterium tuberculosis infections in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:68-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 111]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
72.  Winthrop KL, Kubak BM, Pegues DA, Hufana C, Costamagna P, Desmond E, Sanders C, Shen P, Flores-Ibarra L, Osborne E. Transmission of mycobacterium tuberculosis via lung transplantation. Am J Transplant. 2004;4:1529-1533.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
73.  Kumar D, Budev M, Koval C, Hellinger WC, Gordon SM, Tomford JW. Donor-derived tuberculosis (TB) infection in lung transplant despite following recommended algorithm. Am J Transplant. 2013;13:2225-2226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
74.  Oeltmann JE, Kammerer JS, Pevzner ES, Moonan PK. Tuberculosis and substance abuse in the United States, 1997-2006. Arch Intern Med. 2009;169:189-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 75]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
75.  Cegielski JP, McMurray DN. The relationship between malnutrition and tuberculosis: evidence from studies in humans and experimental animals. Int J Tuberc Lung Dis. 2004;8:286-298.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Guelar A, Gatell JM, Verdejo J, Podzamczer D, Lozano L, Aznar E, Miró JM, Mallolas J, Zamora L, González J. A prospective study of the risk of tuberculosis among HIV-infected patients. AIDS. 1993;7:1345-1349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 120]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
77.  Marks SM, Taylor Z, Qualls NL, Shrestha-Kuwahara RJ, Wilce MA, Nguyen CH. Outcomes of contact investigations of infectious tuberculosis patients. Am J Respir Crit Care Med. 2000;162:2033-2038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 160]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
78.  MacIntyre CR, Kendig N, Kummer L, Birago S, Graham NM. Impact of tuberculosis control measures and crowding on the incidence of tuberculous infection in Maryland prisons. Clin Infect Dis. 1997;24:1060-1067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
79.  Schieffelbein CW, Snider DE. Tuberculosis control among homeless populations. Arch Intern Med. 1988;148:1843-1846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
80.  Cain KP, Haley CA, Armstrong LR, Garman KN, Wells CD, Iademarco MF, Castro KG, Laserson KF. Tuberculosis among foreign-born persons in the United States: achieving tuberculosis elimination. Am J Respir Crit Care Med. 2007;175:75-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 90]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
81.  Zuber PL, McKenna MT, Binkin NJ, Onorato IM, Castro KG. Long-term risk of tuberculosis among foreign-born persons in the United States. JAMA. 1997;278:304-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 102]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
82.  Morris MI, Daly JS, Blumberg E, Kumar D, Sester M, Schluger N, Kim SH, Schwartz BS, Ison MG, Humar A. Diagnosis and management of tuberculosis in transplant donors: a donor-derived infections consensus conference report. Am J Transplant. 2012;12:2288-2300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 93]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
83.  Seem DL, Lee I, Umscheid CA, Kuehnert MJ. Excerpt from PHS guideline for reducing HIV, HBV and HCV transmission through organ transplantation. Am J Transplant. 2013;13:1953-1962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
84.  Lee WM. Hepatitis B virus infection. N Engl J Med. 1997;337:1733-1745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1728]  [Cited by in F6Publishing: 1678]  [Article Influence: 62.1]  [Reference Citation Analysis (0)]
85.  Ueda Y, Marusawa H, Egawa H, Okamoto S, Ogura Y, Oike F, Nishijima N, Takada Y, Uemoto S, Chiba T. De novo activation of HBV with escape mutations from hepatitis B surface antibody after living donor liver transplantation. Antivir Ther. 2011;16:479-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
86.  Chazouilleres O, Mamish D, Kim M, Carey K, Ferrell L, Roberts JP, Ascher NL, Wright TL. “Occult” hepatitis B virus as source of infection in liver transplant recipients. Lancet. 1994;343:142-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 227]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
87.  Uemoto S, Sugiyama K, Marusawa H, Inomata Y, Asonuma K, Egawa H, Kiuchi T, Miyake Y, Tanaka K, Chiba T. Transmission of hepatitis B virus from hepatitis B core antibody-positive donors in living related liver transplants. Transplantation. 1998;65:494-499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 205]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
88.  Douglas DD, Rakela J, Wright TL, Krom RA, Wiesner RH. The clinical course of transplantation-associated de novo hepatitis B infection in the liver transplant recipient. Liver Transpl Surg. 1997;3:105-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 97]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
89.  Fabia R, Levy MF, Crippin J, Tillery W, Netto GJ, Aguanno J, Dysert P, Goldstein RM, Husberg BS, Gonwa TA. De novo hepatitis B infection after liver transplantation: source of disease, incidence, and impact. Liver Transpl Surg. 1998;4:119-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 40]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
90.  Franchello A, Ghisetti V, Marzano A, Romagnoli R, Salizzoni M. Transplantation of hepatitis B surface antigen-positive livers into hepatitis B virus-positive recipients and the role of hepatitis delta coinfection. Liver Transpl. 2005;11:922-928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 50]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
91.  Lauer GM, Walker BD. Hepatitis C virus infection. N Engl J Med. 2001;345:41-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2042]  [Cited by in F6Publishing: 1987]  [Article Influence: 86.4]  [Reference Citation Analysis (0)]
92.  Levitsky J, Doucette K. Viral hepatitis in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:147-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
93.  Terrault N, Roche B, Samuel D. Management of the hepatitis B virus in the liver transplantation setting: a European and an American perspective. Liver Transpl. 2005;11:716-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 127]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
94.  Chung RT, Feng S, Delmonico FL. Approach to the management of allograft recipients following the detection of hepatitis B virus in the prospective organ donor. Am J Transplant. 2001;1:185-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 72]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
95.  Manzarbeitia C, Reich DJ, Ortiz JA, Rothstein KD, Araya VR, Munoz SJ. Safe use of livers from donors with positive hepatitis B core antibody. Liver Transpl. 2002;8:556-561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 75]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
96.  Prakoso E, Strasser SI, Koorey DJ, Verran D, McCaughan GW. Long-term lamivudine monotherapy prevents development of hepatitis B virus infection in hepatitis B surface-antigen negative liver transplant recipients from hepatitis B core-antibody-positive donors. Clin Transplant. 2006;20:369-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
97.  Suehiro T, Shimada M, Kishikawa K, Shimura T, Soejima Y, Yoshizumi T, Hashimoto K, Mochida Y, Maehara Y, Kuwano H. Prevention of hepatitis B virus infection from hepatitis B core antibody-positive donor graft using hepatitis B immune globulin and lamivudine in living donor liver transplantation. Liver Int. 2005;25:1169-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
98.  Saab S, Waterman B, Chi AC, Tong MJ. Comparison of different immunoprophylaxis regimens after liver transplantation with hepatitis B core antibody-positive donors: a systematic review. Liver Transpl. 2010;16:300-307.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Loss GE, Mason AL, Nair S, Blazek J, Farr G, Guo L, Cohen AJ, Eason JD. Does lamivudine prophylaxis eradicate persistent HBV DNA from allografts derived from anti-HBc-positive donors? Liver Transpl. 2003;9:1258-1264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 24]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
100.  Loss GE, Mason AL, Blazek J, Dick D, Lipscomb J, Guo L, Perrillo RP, Eason JD. Transplantation of livers from hbc Ab positive donors into HBc Ab negative recipients: a strategy and preliminary results. Clin Transplant. 2001;15 Suppl 6:55-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
101.  Jain A, Orloff M, Abt P, Kashyap R, Mohanka R, Lansing K, Kelley M, Bozorgzadeh A. Use of hepatitis B core antibody-positive liver allograft in hepatitis C virus-positive and -negative recipients with use of short course of hepatitis B immunoglobulin and Lamivudine. Transplant Proc. 2005;37:3187-3189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
102.  Vizzini G, Gruttadauria S, Volpes R, D’Antoni A, Pietrosi G, Filì D, Petridis I, Pagano D, Tuzzolino F, Santonocito MM. Lamivudine monoprophylaxis for de novo HBV infection in HBsAg-negative recipients with HBcAb-positive liver grafts. Clin Transplant. 2011;25:E77-E81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
103.  Pereira BJ, Wright TL, Schmid CH, Levey AS. A controlled study of hepatitis C transmission by organ transplantation. The New England Organ Bank Hepatitis C Study Group. Lancet. 1995;345:484-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 98]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
104.  Northup PG, Argo CK, Nguyen DT, McBride MA, Kumer SC, Schmitt TM, Pruett TL. Liver allografts from hepatitis C positive donors can offer good outcomes in hepatitis C positive recipients: a US National Transplant Registry analysis. Transpl Int. 2010;23:1038-1044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 92]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
105.  Delladetsima I, Psichogiou M, Sypsa V, Psimenou E, Kostakis A, Hatzakis A, Boletis JN. The course of hepatitis C virus infection in pretransplantation anti-hepatitis C virus-negative renal transplant recipients: a retrospective follow-up study. Am J Kidney Dis. 2006;47:309-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
106.  Maluf DG, Archer KJ, Mas VR. Kidney grafts from HCV-positive donors: advantages and disadvantages. Transplant Proc. 2010;42:2436-2446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
107.  Bucci JR, Matsumoto CS, Swanson SJ, Agodoa LY, Holtzmuller KC, Peters TG, Abbott KC. Donor hepatitis C seropositivity: clinical correlates and effect on early graft and patient survival in adult cadaveric kidney transplantation. J Am Soc Nephrol. 2002;13:2974-2982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 75]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
108.  Kucirka LM, Singer AL, Ros RL, Montgomery RA, Dagher NN, Segev DL. Underutilization of hepatitis C-positive kidneys for hepatitis C-positive recipients. Am J Transplant. 2010;10:1238-1246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 134]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
109.  Manuel O, Estabrook M. RNA respiratory viruses in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:212-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
110.  Couch RB, Englund JA, Whimbey E. Respiratory viral infections in immunocompetent and immunocompromised persons. Am J Med. 1997;102:2-9; discussion 25-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 217]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
111.  Ison MG, Gubareva LV, Atmar RL, Treanor J, Hayden FG. Recovery of drug-resistant influenza virus from immunocompromised patients: a case series. J Infect Dis. 2006;193:760-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 214]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
112.  Billings JL, Hertz MI, Savik K, Wendt CH. Respiratory viruses and chronic rejection in lung transplant recipients. J Heart Lung Transplant. 2002;21:559-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 164]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
113.  Khalifah AP, Hachem RR, Chakinala MM, Schechtman KB, Patterson GA, Schuster DP, Mohanakumar T, Trulock EP, Walter MJ. Respiratory viral infections are a distinct risk for bronchiolitis obliterans syndrome and death. Am J Respir Crit Care Med. 2004;170:181-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 251]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
114.  Kumar D, Erdman D, Keshavjee S, Peret T, Tellier R, Hadjiliadis D, Johnson G, Ayers M, Siegal D, Humar A. Clinical impact of community-acquired respiratory viruses on bronchiolitis obliterans after lung transplant. Am J Transplant. 2005;5:2031-2036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 185]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
115.  Meylan PR, Aubert JD, Kaiser L. Influenza transmission to recipient through lung transplantation. Transpl Infect Dis. 2007;9:55-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
116.  Le Page AK, Kainer G, Glanville AR, Tu E, Bhonagiri D, Rawlinson WD. Influenza B virus transmission in recipients of kidney and lung transplants from an infected donor. Transplantation. 2010;90:99-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
117.  Kozlowski T, Nickeleit V, Andreoni K. Donor-transmitted adenovirus infection causing kidney allograft nephritis and graft loss. Transpl Infect Dis. 2011;13:168-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
118.  Kumar D, Morris MI, Kotton CN, Fischer SA, Michaels MG, Allen U, Blumberg EA, Green M, Humar A, Ison MG. Guidance on novel influenza A/H1N1 in solid organ transplant recipients. Am J Transplant. 2010;10:18-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 110]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
119.  Florescu DF, Hoffman JA. Adenovirus in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:206-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
120.  Shames BD, D’Alessandro AM, Sollinger HW. Human T-cell lymphotrophic virus infection in organ donors: a need to reassess policy? Am J Transplant. 2002;2:658-663.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
121.  Armstrong MJ, Corbett C, Rowe IA, Taylor GP, Neuberger JM. HTLV-1 in solid-organ transplantation: current challenges and future management strategies. Transplantation. 2012;94:1075-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 35]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
122.  Toro C, Rodés B, Poveda E, Soriano V. Rapid development of subacute myelopathy in three organ transplant recipients after transmission of human T-cell lymphotropic virus type I from a single donor. Transplantation. 2003;75:102-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 101]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
123.  Kaul DR, Taranto S, Alexander C, Covington S, Marvin M, Nowicki M, Orlowski J, Pancoska C, Pruett TL, Ison MG. Donor screening for human T-cell lymphotrophic virus 1/2: changing paradigms for changing testing capacity. Am J Transplant. 2010;10:207-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
124.  Waggoner JJ, Soda EA, Deresinski S. Rare and emerging viral infections in transplant recipients. Clin Infect Dis. 2013;57:1182-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
125.  Rabe IB, Schwartz BS, Farnon EC, Josephson SA, Webber AB, Roberts JP, de Mattos AM, Gallay BJ, van Slyck S, Messenger SL. Fatal transplant-associated west nile virus encephalitis and public health investigation-california, 2010. Transplantation. 2013;96:463-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
126.  Singh N, Levi ME. Arenavirus and West Nile virus in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:361-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
127.  Singh N, Huprikar S, Burdette SD, Morris MI, Blair JE, Wheat LJ. Donor-derived fungal infections in organ transplant recipients: guidelines of the American Society of Transplantation, infectious diseases community of practice. Am J Transplant. 2012;12:2414-2428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 119]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
128.  Canaud G, Timsit MO, Zuber J, Bougnoux ME, Méjean A, Thervet E, Snanoudj R, Sberro R, Martinez F, Legendre C. Early conservative intervention for candida contamination of preservative fluid without allograft nephrectomy. Nephrol Dial Transplant. 2009;24:1325-1327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
129.  Koo S, Kubiak DW, Issa NC, Dietzek A, Boukedes S, Camp PC, Goldberg HJ, Baden LR, Fuhlbrigge AL, Marty FM. A targeted peritransplant antifungal strategy for the prevention of invasive fungal disease after lung transplantation: a sequential cohort analysis. Transplantation. 2012;94:281-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
130.  Laouad I, Buchler M, Noel C, Sadek T, Maazouz H, Westeel PF, Lebranchu Y. Renal artery aneurysm secondary to Candida albicans in four kidney allograft recipients. Transplant Proc. 2005;37:2834-2836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 49]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
131.  Sun HY, Wagener MM, Singh N. Cryptococcosis in solid-organ, hematopoietic stem cell, and tissue transplant recipients: evidence-based evolving trends. Clin Infect Dis. 2009;48:1566-1576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 68]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
132.  Saha DC, Goldman DL, Shao X, Casadevall A, Husain S, Limaye AP, Lyon M, Somani J, Pursell K, Pruett TL. Serologic evidence for reactivation of cryptococcosis in solid-organ transplant recipients. Clin Vaccine Immunol. 2007;14:1550-1554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 105]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
133.  Baddley JW, Schain DC, Gupte AA, Lodhi SA, Kayler LK, Frade JP, Lockhart SR, Chiller T, Bynon JS, Bower WA. Transmission of Cryptococcus neoformans by Organ Transplantation. Clin Infect Dis. 2011;52:e94-e98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 75]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
134.  Pappas PG, Perfect JR, Cloud GA, Larsen RA, Pankey GA, Lancaster DJ, Henderson H, Kauffman CA, Haas DW, Saccente M. Cryptococcosis in human immunodeficiency virus-negative patients in the era of effective azole therapy. Clin Infect Dis. 2001;33:690-699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 446]  [Cited by in F6Publishing: 416]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
135.  Perfect JR, Dismukes WE, Dromer F, Goldman DL, Graybill JR, Hamill RJ, Harrison TS, Larsen RA, Lortholary O, Nguyen MH. Clinical practice guidelines for the management of cryptococcal disease: 2010 update by the infectious diseases society of america. Clin Infect Dis. 2010;50:291-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1913]  [Cited by in F6Publishing: 1678]  [Article Influence: 119.9]  [Reference Citation Analysis (0)]
136.  Cuellar-Rodriguez J, Avery RK, Lard M, Budev M, Gordon SM, Shrestha NK, van Duin D, Oethinger M, Mawhorter SD. Histoplasmosis in solid organ transplant recipients: 10 years of experience at a large transplant center in an endemic area. Clin Infect Dis. 2009;49:710-716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 127]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
137.  Blair JE, Logan JL. Coccidioidomycosis in solid organ transplantation. Clin Infect Dis. 2001;33:1536-1544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 137]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
138.  Vucicevic D, Carey EJ, Blair JE. Coccidioidomycosis in liver transplant recipients in an endemic area. Am J Transplant. 2011;11:111-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
139.  Miller MB, Hendren R, Gilligan PH. Posttransplantation disseminated coccidioidomycosis acquired from donor lungs. J Clin Microbiol. 2004;42:2347-2349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
140.  Miller R, Assi M. Endemic fungal infections in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:250-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
141.  Wright PW, Pappagianis D, Wilson M, Louro A, Moser SA, Komatsu K, Pappas PG. Donor-related coccidioidomycosis in organ transplant recipients. Clin Infect Dis. 2003;37:1265-1269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 78]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
142.  Bern C, Montgomery SP, Herwaldt BL, Rassi A, Marin-Neto JA, Dantas RO, Maguire JH, Acquatella H, Morillo C, Kirchhoff LV. Evaluation and treatment of chagas disease in the United States: a systematic review. JAMA. 2007;298:2171-2181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 545]  [Cited by in F6Publishing: 478]  [Article Influence: 28.1]  [Reference Citation Analysis (0)]
143.  Vincenzi R, Neto JS, Fonseca EA, Pugliese V, Leite KR, Benavides MR, Cândido HL, Porta G, Miura IK, Pugliese R. Schistosoma mansoni infection in the liver graft: The impact on donor and recipient outcomes after transplantation. Liver Transpl. 2011;17:1299-1303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
144.  Roseman DA, Kabbani D, Kwah J, Bird D, Ingalls R, Gautam A, Nuhn M, Francis JM. Strongyloides stercoralis transmission by kidney transplantation in two recipients from a common donor. Am J Transplant. 2013;13:2483-2486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
145.  Mossad SB. Region-specific challenges for minimizing endemic donor-transmitted infections. Liver Transpl. 2011;17:1241-1243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
146.  Schwartz BS, Mawhorter SD. Parasitic infections in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:280-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 122]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
147.  Coura JR, Borges-Pereira J. Chagas disease. What is known and what should be improved: a systemic review. Rev Soc Bras Med Trop. 2012;45:286-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 89]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
148.  Chin-Hong PV, Schwartz BS, Bern C, Montgomery SP, Kontak S, Kubak B, Morris MI, Nowicki M, Wright C, Ison MG. Screening and treatment of chagas disease in organ transplant recipients in the United States: recommendations from the chagas in transplant working group. Am J Transplant. 2011;11:672-680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 116]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
149.  Schwartz BS, Paster M, Ison MG, Chin-Hong PV. Organ donor screening practices for Trypanosoma cruzi infection among US Organ Procurement Organizations. Am J Transplant. 2011;11:848-851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
150.  Machado CM, Levi JE. Transplant-associated and blood transfusion-associated tropical and parasitic infections. Infect Dis Clin North Am. 2012;26:225-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
151.  Rassi A, Rassi A, Marcondes de Rezende J. American trypanosomiasis (Chagas disease). Infect Dis Clin North Am. 2012;26:275-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in F6Publishing: 357]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
152.  Huprikar S, Bosserman E, Patel G, Moore A, Pinney S, Anyanwu A, Neofytos D, Ketterer D, Striker R, Silveira F. Donor-derived Trypanosoma cruzi infection in solid organ recipients in the United States, 2001-2011. Am J Transplant. 2013;13:2418-2425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 66]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
153.  Boyarsky BJ, Hall EC, Singer AL, Montgomery RA, Gebo KA, Segev DL. Estimating the potential pool of HIV-infected deceased organ donors in the United States. Am J Transplant. 2011;11:1209-1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 85]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]