case report

Isolated Primary Central Nervous System Lymphoma Renal Transplant Treated with Surgical and Pharmacologic Intervention: A Case Report and Review of Literature

Ali Eltatawy*, Meera Raghavan, Hayder Abdulwahid, Dalia Ibrahim, Robert Booth, Jorge Ortiz, Robert Mrak

Department of Pathology, University of Toledo Medical Center, Toledo Ohio, USA

*Corresponding author: Ali Eltatawy, Department of Pathology, University of Toledo Medical Center, Toledo Ohio, USA. Email: Ali.el2@rockets.utoledo.edu

Received Date: 20 September, 2019; Accepted Date: 15 October, 2019; Published Date: 21 October, 2019

Citation: Eltatawy A, Raghavan M, Abdulwahid H, Ibrahim D, Booth R, et al. (2019) Isolated Primary Central Nervous System Lymphoma Renal Transplant Treated with Surgical and Pharmacologic Intervention: A Case Report and Review of Literature. J Surg 4: 1255. DOI: 10.29011/2575-9760.001255

Abstract

We present a rare variant of Post–Transplant Lymphoproliferative Disorder (PTLD); Primary Central Nervous System (PCNS) lymphoma, in a demographic not consistent with previous reports. A 51-year-old non-Hispanic white female with a history of adult polycystic kidney disease, status-post two renal transplants presented five years after her 2nd transplant with new onset neurological symptoms. Radiography confirmed a single 11 x 10 mm left posterior parietal lobe ring-enhancing mass with surrounding edema and abnormal spectroscopy highly suggestive of malignancy. Histomorphology and immunohistochemical findings were consistent with WHO 2008 classification for Diffuse Large B-Cell Lymphoma (DLBCL). Treatment included surgical resection and high dose methotrexate instead of the recommended initial therapy. Primary central nervous system lymphoma is a rare presentation of post-transplant lymphoproliferative disorder whose clinical attributes are not well known to many clinicians. As the number of successful transplants increases, the likelihood of encountering Primary Central Nervous System Post-Transplant Lymphoproliferative Disorder (PCNS-PTLD) increases. It is important for clinicians to be aware of the current literature, risk factors, variance in presentation, and options for management of this rare disease.

Keywords

Cytomegalovirus (MeSH ID: D003587); Human Herpes virus 4 (MeSH ID: D004854), Lymphoproliferative disorders (MeSH ID: D008232); Renal transplantation (MeSH ID D014180)

Introduction

Post–Transplant Lymphoproliferative Disorder (PTLD) is a lymphoid proliferation in the setting of immunosuppression, especially in solid organ or allogenic hematopoietic cell transplantation [1]. It is the most common malignancy complicating solid organ transplantation after non-melanoma skin cancer and in situ cervical cancer [2,3]. PTLD usually (>80%) presents in the first year post-transplant with lymphoid proliferation in the allograft tissue, in blood or in adjacent organs [4-6]. However, approximately 5-15% of PTLD present as Primary Central Nervous System Post-Transplant Lymphoproliferative Disorders (PCNS-PTLD) [6,7](Table 1).

This rare presentation portends a poorer prognosis, with an estimated three-year survival rate of 32–38% [4,6]. The World Health Organization (WHO) divided PTLD into three general types (Figure 1). PCNS-PTLD histology is most often monomorphic with large B-cell morphology [4,15,16]. Monomorphic PTLD is a monoclonal lymphoid proliferation that meets criteria for one of the B-cell or T/NK cell lymphomas [17]. A combination of Magnetic Resonance Imaging (MRI) and histopathology are the gold standard in diagnosis [18,19].

As transplantation and immunosuppression advance, the number of successful transplants exponentially increases, and with it the incidence of PCNS-PTLD. However, there are no current standards for screening or treatment of patients with PCNS-PTLD. Therefore, we present here a case report and review of literature to distinguish the differences between systemic and PCNS PTLD, and distill the growing evidence for management.

Case Report

Our patient is a 51-year-old non-Hispanic white female with a history of adult polycystic kidney disease, status-post two renal transplants. She underwent a deceased donor renal transplant in 2005, at which time she was EBV and CMV positive. Immunosuppressive therapy included mycophenolate mofetil 1000 mg bid, cyclosporine (ranging from 200 mg bid to 150 mg bid over 3 years), prednisone tapering (30 mg to 10 mg over 1 year), and antiviral therapy included valgancyclovir (3 months). The transplant failed within a year due to medication non-adherence, and she resumed dialysis until her second transplant in 2014. The recipient and second donor were both CMV and EBV positive. Her initial immunosuppressive therapy included alemtuzumab (monoclonal antibody) induction, tacrolimus 1.5 to 2 mg bid., mycophenolate 720 mg bid., in addition to antiviral valgancyclovir 450 mg. Due to rejection, seven doses of thymoglobin, pulse steroids and eventually daily prednisone 10 mg were prescribed.

Intermittent numbness in the right arm was reported in November 2018. She could not tolerate an MRI due to anxiety. CT brain without contrast revealed a subtle hypo-attenuating focus within the left parietal lobe designated “most likely a small remote infarct” (Figure 2). When neurological symptoms progressed further, sedation assisted brain MRI with contrast revealed a left parietal mass demonstrating extensive enhancement. Radiography confirmed an 11 x 10 mm left posterior parietal lobe ring-enhancing mass with surrounding edema and spectroscopy was highly suggestive of malignancy (Figure 2). Neurosurgery biopsied and resected the tumor.

The mass was described as a spherical homogenous lesion measuring 40 x 20 x 10 mm. Histopathological examination of multiple representative sections revealed sheets of large atypical cells and normal appearing lymphocytes (Figure 3). Ancillary immune histochemical staining was performed to profile the tumor (Figure 4). The atypical cells were negative for GFAP. Strong positivity for both CD20 and CD30 confirmed B-cell origin. Immunostains were consistent with large B-cell lymphoma of non-germinal center type; CD10 was negative, while both BCL6 and Mum1 were positive. Scattered T-cells in the background stained positively for CD3. Immunohistochemical analysis revealed a lack of Myc and p53 expression. EBV in situ hybridization was strongly positive in the lymphocytes. B-cell clonality determination by PCR was performed at an outside facility and demonstrated positivity for IgH and IgK gene rearrangement. The final pathologic diagnosis was large B cell CNS lymphoma according to WHO 2008 classifications.

The post-surgical treatment consists of high-dose methotrexate infusion and leucovorin treatments. As anticipated, increased creatinine was reported after methotrexate administration. Her immunosuppressive therapy was switched to an mTOR based regimen.

Discussion

Epidemiology

The incidence of PCNS-PTLD varies with age, organ type transplanted, ethnicity, and type of immunosuppression. This non-Hispanic white woman in her 6th decade of life presented with neurologic symptoms approximately 13 years following her first renal transplant and approximately 4 years after her second renal transplant. The median age of PCNS-PTLD diagnosis is between 48-60 years [2,8] and disease typically presents 4-5 years post-transplant [4,5,12,13]. Both systemic and PCNS PTLD are more common in non-Hispanic white individuals, while PCNS-PTLD has a 2 times higher incidence in Asian/pacific islanders than non-Hispanic white patients [11] (Table 1). Unlike systemic PTLD [9,13,20], PCNS-PTLD is commonly associated with renal transplants [4,15,11] (Table 2).

EBV and CMV

EBV, CMV, and other viruses have an association with the development of PTLD [4-6,21,22]. However, there are inconsistent screening guidelines for early PTLD based on these viruses and there are no guidelines for PCNS-PTLD or late-presenting PTLD. Viral screening was positive for EBV and CMV at her first transplantation. Only the donor of her second renal transplant was positive for EBV and CMV. EBV is able to induce uncontrolled proliferation of B cells, and is theorized to induce PTLD [8,23]. Evens et al. reported 97% of PCNS-PTLD tumors positive for EBV virus [4]. Most cases of systemic PTLD and PCNS-PTLD are related to the presence of EBV in either the recipient or the transplanted organ [4,8,11] (Table 2).

However, Mahale, et al. reports a higher incidence of PCNS-PTLD in transplant recipients who are seronegative for EBV and at risk for primary infection following transplant [11]. Some have suggested monitoring for EBV DNA in the peripheral blood in the early transplant period in high risk seronegative patients [30]. Unfortunately, these guidelines fail to account for individuals presenting with late onset PTLD, which is often EBV negative, and miss approximately half of EBV-positive cases with no detectable EBV in the peripheral blood [8,31]. Our PCR results demonstrated positive serum EBV followed by negative serum EBV within two months of each other, further supporting the notion that serum EBV is not a reliable screening method. Screening Cerebrospinal Fluid (CSF) for EBV to monitor for PCNS-PTLD also proved futile by Evens et al, as only 10% of PCNS-PTLD were detectable in CSF [4]. In fact, the presence of EBV DNA in CSF due to PCNS-PTLD was lower than EBV DNA levels in encephalitis or brain abscess [32]. Ancillary testing was positive for EBV on in-situ hybridization and negative for CSF involvement despite having isolated, aggressive PCNS-PTLD. Other viruses, such as human T-cell lymphotropic virus, human herpesvirus 8, CMV, simian virus 40 and hepatitis C have been proposed to increase the incidence of PTLD although they were not observed in this case [6,33].

Clinical Presentation

The clinical presentation of PCNS-PTLD can be remarkably variable, corresponding to the specific region (s) of the brain involved [6,19,34]. Certain lab findings such as anemia, leukopenia, thrombocytopenia, elevated LDH, elevated calcium ion in blood, or hyperuricemia may be useful in narrowing the diagnosis and prognostication [8]. Significant lab values in this case included elevated serum creatinine, worsening thrombocytopenia, low haptoglobin, and elevated LDH.

Diagnostic imaging

In post-transplant patients who fit the epidemiologic profile for development of PCNS-PTLD and have supportive clinical or laboratory findings, prompt imaging with CT and early follow-up MRI should be conducted as these lesions are often aggressive [11]. PCNS-PTLD most frequently presents as multiple ring-enhancing lesions that are typically supratentorial, lobar or paraventricular, and involve subcortical white matter or basal ganglia [4,19]. However, radiography exhibited a larger, single supratentorial ring-enhancing lesion of the peripheral sub cortex inconsistent with the literature.

Histology

Ultimately, a definitive diagnosis of PCNS-PTLD can only be accomplished by histologic assessment to profile the lesion and rule out opportunistic infections that often present with similar imaging findings [19]. Immunohistochemical examination will typically demonstrate EBV positivity, lack of p53 expression, lack of Myc expression, and numerous background T lymphocytes [14] which is consistent with our pathologic findings (Figure 4). Isolated PCNS-PTLD is most frequently a monomorphic B cell lesion (2008 WHO classification) which is typically more aggressive [4,11,15,16].

Immunosuppression

Certain drug therapies may be risk modifying for PCNS-PTLD development based on their mechanism of action and degree of immunosuppression [3]. After her first renal transplant, she received Mycophenolate Mofetil (MMF), cyclosporine, and prednisone as part of her standard immunosuppressive therapy to prevent allograft rejection. MMF has been associated with increased risk of PCNS-PTLD [15], particularly when administered without calcineurin inhibitors (cyclosporine, tacrolimus) [8,15]. Calcineurin inhibitors are protective against PCNS-PTLD, despite an association with increased risk for systemic PTLD [5,9,10,25], (Table 2). These calcineurin inhibitors protect against the increased risk of PCNS-PTLD conferred by mycophenolate mofetil [5].

Following the second renal transplant, she received alemtuzumab induction, mycophenolate mofetil, tacrolimus and several doses of thymoglobulin for rejection. According to Oliver et al. 2017, Alemtuzumab induction is not associated with increased risk of systemic PTLD in kidney transplantation [35]. However, it is associated with increased incidence of PCNS-PTLD [8,11]. The use of thymoglobulin has also been associated with increased incidence of PCNS-PTLD [11]. After surgery the immunosuppressive regimen was switched to mTOR based regimen. While there are conflicting data regarding whether or not mTOR inhibitors impart an increased risk of PCNS-PTLD, sirolimus increases risk of PTLD twofold [24,28].

Management

Owing to the rarity of PCNS-PTLD and its aggressive clinical course, there is little published data regarding treatment. Options commonly proposed include reduction or withdrawal of immunosuppression, whole brain radiotherapy, rituximab and high dose methotrexate [4,8,36-38]. Unlike monomorphic PTLD [39], the recommended initial step in treatment of PCNS-PTLD is reduction of immunotherapy [4]. This must be supplemented by radiation or another pharmaceutical first line therapy [4]. Withdrawal of immunosuppression should be balanced against the risk of transplant rejection. There is one case report by Yaginuma et al. of successful treatment of isolated PCNS-PTLD by whole brain radiotherapy alone [38]. However, other sources suggest radiotherapy may not be as effective on late onset PCNS-PTLD as early systemic PTLD [8]. Some case reports demonstrate complete resolution of PCNS-PTLD after treatment with rituximab, an anti-CD20 antibody [1,13]. Although anti-CD20 antibody has been proven effective against systemic PTLD, it is known to have difficulty passing through the blood-brain barrier [38], making it unlikely to be effective as the sole treatment of PCNS-PTLD. High dose methotrexate has been effective as an isolated therapy for PCNS-PTLD in several cases [23,37,40]. Due to the isolated nature of the brain lesion, surgical excision was considered the best course of action and was followed by administration of high dose methotrexate. Leucovorin was also incorporated into her treatment regimen.

Other treatments described in the literature have demonstrated mixed results, including laboratory treated cytotoxic T-cell lymphocyte infusion with craniotomy, temozolomide, surgical resection and antiviral medication [8,41,42]. Factors such as age, timing of disease onset, laboratory findings and histology may be useful in prognostication. As reported by Caillard, et al. CNS localization is an independent prognostic indicator of poor survival [43]. Age >55 years at diagnosis, late onset PTLD, high LDH and creatinine levels, widespread PTLD, T cell lymphoma and monomorphic histology are also associated with poor prognosis [43]. The most dominant prognostic factor reported by Evens et al. was the lack of response to first line therapy [4]. The clinician should start with reduction of immunotherapy as an initial step in treatment and supplement with radiotherapy and rituximab. When the response is limited or isolated lesion is excised, one should discontinue that regimen and treat with high dose methotrexate.

Conclusion

PCNS-PTLD is a rare complication of solid organ transplantation after receiving immunosuppressive therapy for several years [1]. Historically this diagnosis has portended a dismal prognosis with an estimated three-year survival rate of 32–38% [4,6]. Therefore, aggressive diagnosis and low threshold for imaging is important, especially in those who fit the epidemiologic and clinical profile. The rarity of this disease has impeded development of guidelines for prevention, screening, or management. Scattered case reports in the literature were enhanced by a large (84 patients) series reported in 2013 by Evens et al. and [1,4]. Over the past decade the number of successful transplants performed has grown, and so has the incidence of PCNS-PTLD, enabling more actionable analysis. This case report contributes case-based evidence to a limited, albeit growing body of literature on PCNS-PTLD and highlights a novel approach in successful treatment incorporating both surgical and pharmacologic intervention. It is important that, as the literature grows, the medical communities update guidelines, identify outliers and pursue trials to better understand the many factors that determine disease progression and prognosis.


Figure 1: WHO Classification of Post-Transplant Lymphoproliferative Disorder (PTLD).



Figure 2: CT without contrast and MRI. A: CT axial B: CT coronal C: CT sagittal D: MRI T1 axial E: MR T1 coronal F: MRI T1 sagittal. A-C: CT without contrast exhibited a subtle hypo-attenuating focus within the left parietal lobe, designated “small remote infarct” (red arrow). This is most visible on the sagittal view C. D-F: MRI confirmed a left posterior parietal lobe ring-enhancing mass, measuring 11 x 10 mm, with surrounding edema.



Figure 3: Brain Biopsy A: Tissue section; low power view displaying infiltration of the brain tissue by atypical lymphoid cells (Hematoxylin-eosin stain, original magnification X 200). B: Tissue section; higher power view displaying large atypical lymphoid cells to small normal appearing lymphocytes (Hematoxylin-eosin stain, original magnification X 400).



Figure 4: Immunohistochemistry A: CD20 stain of brain tissue biopsy reveals CD20+ B-cell infiltrates. B: many of the large cells were also strongly positive for CD30. C: CD3 stain suggests increased presence of scattered T-cells in the background. D: The atypical cells were negative for GFAP. E, BCL6 staining was positive. F, Mum1 staining was positive. Many cells also stain positive for EBV-encoded RNA (not depicted).

Risk Factor

Systemic PTLD, early presentation <1 year

Systemic PTLD, Late presentation, >1 years

PCNS-PTLD

Study Reference

Age at transplantation

 <25 yrs, mostly children.

Bimodal <20 years old and >50 years old

Median age of 60 years

Systemic [8-10] PCNS

Ethnicity

Non-hispanic white

Non-hispanic white

Asian/pacific islander = 2x incidence than non-hispanic white

Systemic [4-6] PCNS [11]

Time from transplant

60-80% within 1.5 yrs

30% within 1-5 years. (EBV negative median is 4.5 yrs)

Median= 54 months Typically 4-5 years

Systemic [4-6] PCNS [4,11-13]

EBV status on histology at diagnosis

70-95% positive

38% of late were EBV negative.

>90% positive

Systemic [5] PCNS [14]

Note: PTLD= Post-Transplant Lymphoproliferative Disorder, PCNS= Primary Central Nervous System, EBV= Ebstein-Barr Virus


Table 1: Epidemiology and Risk PTLD VS PCNS-PTLD.

Risk Factor

Systemic PTLD

PCNS-PTLD

Study Reference

De novo CMV infection

 

 

 

CMV status of recipient at transplant

Seronegativity 2-4x avg risk

No recent data

[4-6]

CMV status of donor

CMV-negative receiving CMV positive organ 4-6x more likely

No recent data

[4-6]

EBV status

 

 

 

EBV status of recipient at transplant

Seronegativity 10-75x incidence over that of EBV seropositive

Seronegative 2x incidence

[11,21,22]

EBV status of donor

No recent data

No recent data

 

Type of transplant treatment

 

 

 

Calcineurin inhibitors

2x-5x increased risk with prolonged high doses of tacrolimus vs cyclosporine. Cyclosporine increases in higher doses (>6.6mg/kg/day)

Does not significantly increase risk. If added to MMF, reduces the risk association to only 18x fold increased risk.

[10,15,24,25]

MMF without calcineurin inhibitor

Lower risk of PTLD

118x higher odds of PCNS.

[15,24,26,27]

mTOR inhibitors

Sirolimus, 1-2x increased risk

Conflicting data

[24,28]

Polyclonal antibodies: ATG (Antithymocyte globulin)

3-4x increased risk in the 1st year post transplant

2.03x incidence

[11,24,10]

Monoclonal antibodies: OKT3(Anti-CD3 monoclonal antibody)

3-4x increased risk in the 1st year post transplant with high cumulative doses (contested in more recent studies)

1.83x incidence

[11,24,10,29]

IL-2 receptor antibodies

No increased risk

No recent data

[10,24]

Anti-CD52 antibody

No increased risk with alemtuzumab

3.12x aiRR

 [8,11,28]

Type of Organ Transplant

 

 

 

Small bowel

6-18x incidence of kidney

No recent data

[9]

Lung

2-3x incidence of kidney

0.5x incidence of kidney

[9,11]

Multiple organ transplant

1-2x incidence of kidney

2.45x incidence of just 1 kidney transplant

[9-11]

Liver

Same incidence as kidney

0.5x incidence ofkidney

[9,11]

Kidney

1%-3% of all transplants, lowest of all solid organ transplants

58-79% of All PCNS-PTLD

 [4,9,11]

Organ involved

More commonly arise in graft itself, GI or extranodal sites.

5%-25% of all PTLD cases are primary CNS, 11.77 % of PTLD folowing kidney transplant are PCNS

[3,11,15]

Type of lymphoma most associated

Monomorphic PTLD, B-cell

83% monomorphic PTLD, B-cell

[6]

Note: Airr = Adjusted Incidence Ratio, MMF= Mycophenolate Mofetil, PTLD= Post-Transplant Lymphoproliferative Disorder, PCNS= Primary Central Nervous System, EBV= Ebstein-Barr Virus, CMV= Cytomegalovirus, Avg= Average


Table 2: Risk factors PTLD VS PCNS-PTLD.

References

  1. Morris J, Smith C, Streicher A, Magnuson A, Newman S, et al. (2017) A Rare Presentation of Isolated CNS Posttransplantation Lymphoproliferative Disorder. Case Rep Oncol Med 2017: 7269147.
  2. Ghobrial IM, Habermann TM, Maurer MJ, Geyer SM, Ristow KM, et al. (2005) Prognostic analysis for survival in adult solid organ transplant recipients with post-transplantation lymphoproliferative disorders. J Clin Oncol 23: 7574-7582.
  3. Bagg A, Dunphy CH (2013) Immunosuppressive and immunomodulatory therapy-associated lymphoproliferative disorders. Seminars in Diagnostic Pathology 30: 102-112.
  4. Evens AM, Choquet S, Kroll-Desrosiers AR, Jagadeesh D, Smith SM, et al. (2013) Primary CNS Posttransplant Lymphoproliferative Disease (PTLD): An International Report of 84 Cases in the Modern Era 13: 1512-1522.
  5. Morton M, Coupes B, Roberts SA, Klapper PE, Byers RJ, et al. (2013) Epidemiology of posttransplantation lymphoproliferative disorder in adult renal transplant recipients. Transplantation 95: 470-478.
  6. Cavaliere R, Gina Petroni, Maria B. Lopes, David Schiff, The International Primary Central Nervous System Lymphoma Collaborative Group (2010) Primary central nervous system post-transplantation lymphoproliferative disorder. Cancer 116: 863-870.
  7. Kempf C, Tinguely M, Rushing EJ (2013) Posttransplant Lymphoproliferative Disorder of the Central Nervous System. Pathobiology 80: 310-318.
  8. Velvet AJJ, Bhutani S, Papachristos S, Dwivedi R, Picton M, et al. (2019) A single-center experience of post-transplant lymphomas involving the central nervous system with a review of current literature. Oncotarget 10: 437-448.
  9. Taylor AL, Marcus R, Bradley JA (2005) Post-transplant lymphoproliferative disorders (PTLD) after solid organ transplantation. Crit Rev Oncol Hematol 56: 155-167.
  10. Opelz G, Döhler B (2004) Lymphomas After Solid Organ Transplantation: A Collaborative Transplant Study Report. Am J Transplant 4: 222-230.
  11. Mahale P, Shiels MS, Lynch CF, Engels EA (2018) Incidence and outcomes of primary central nervous system lymphoma in solid organ transplant recipients. American journal of transplantation 18: 453-461.
  12. Tsao L, His ED (2007) The clinicopathologic spectrum of posttransplantation lymphoproliferative disorders. Arch Pathol Lab Med 131: 1209-1218.
  13. Gottschalk S, Rooney CM, Heslop HE (2005) Post-transplant lymphoproliferative disorders. Annu Rev Med 56: 29-44.
  14. Sundin A, Grzywacz BJ, Yohe S, Linden MA, Courville EL (2017) B-cell posttransplant lymphoproliferative disorder isolated to the central nervous system is Epstein-Barr virus positive and lacks p53 and Myc expression by immunohistochemistry. Human Pathology 61: 140-147.
  15. Crane GM, Powell H, Kostadinov R, Rocafort PT, Rifkin DE, et al. (2015) Primary CNS lymphoproliferative disease, mycophenolate and calcineurin inhibitor usage. Oncotarget 6: 33849-33866.
  16. Evens AM, Roy R, Sterrenberg D, Moll MZ, Chadburn A, et al. (2010) Post-transplantation lymphoproliferative disorders: diagnosis, prognosis, and current approaches to therapy. Curr Oncol Rep 12: 383-394.
  17. Swerdlow SH, CE, Harris NL, Jaffe ES, Pileri SA, et al. (2008) WHO Classification of Tumors of Haematopoietic and Lymphoid Tissues IARC: Lyuon 2008.
  18. Twombley K, Pokala H, Ardura MI, Harker-Murray P, Johnson-Welch SF, et al. (2012) Intraventricular rituximab and systemic chemotherapy for treatment of central nervous system post-transplant lymphoproliferative disorder after kidney transplantation 16: 201-209.
  19. Lake W, Chang JE, Kennedy T, Morgan A, Salamat S, et al. (2013) A case series of primary central nervous system posttransplantation lymphoproliferative disorder: imaging and clinical characteristics. Neurosurgery 72: 960-970.
  20. Faull RJ, Hollett P, McDonald SP (2005) Lymphoproliferative disease after renal transplantation in Australia and New Zealand. Transplantation 80: 193-197.
  21. Walker RC, Marshall WF, Strickler JG, Wiesner RH, Velosa JA, et al. (1995) Pretransplantation assessment of the risk of lymphoproliferative disorder. Clin Infect Dis 20: 1346-1353.
  22. Cockfield SM (2001) Identifying the patient at risk for post-transplant lymphoproliferative disorder. Transpl Infect Dis 3: 70-78.
  23. Hatton O, Martinez OM, Esquivel CO (2012) Emerging therapeutic strategies for Epstein-Barr virus+ post-transplant lymphoproliferative disorder. Pediatr Transplant 16: 220-229.
  24. Caillard S, Dharnidharka V, Agodoa L, Bohen E, Abbott K (2005) Posttransplant lymphoproliferative disorders after renal transplantation in the United States in era of modern immunosuppression. Transplantation 80: 1233-1243.
  25. Younes BS, McDiarmid SV, Martin MG, Vargas JH, Goss JA, et al. (2000) The effect of immunosuppression on posttransplant lymphoproliferative disease in pediatric liver transplant patients. Transplantation 70: 94-99.
  26. Robson R, Cecka JM, Opelz G, Budde M, Sacks S (2005) Prospective registry-based observational cohort study of the long-term risk of malignancies in renal transplant patients treated with mycophenolate mofetil. Am J Transplant 5: 2954-2960.
  27. Funch DP, Ko HH, Travasso J, Brady J, Kew CE, et al. (2005) Posttransplant lymphoproliferative disorder among renal transplant patients in relation to the use of mycophenolate mofetil. Transplantation 80: 1174-1180.
  28. Kirk AD, Cherikh WS, Ring M, Burke G, Kaufman D, et al. (2007) Dissociation of depletional induction and posttransplant lymphoproliferative disease in kidney recipients treated with alemtuzumab. Am J Transplant 7: 2619-2625.
  29. Swinnen LJ, Costanzo-Nordin MR, Fisher SG, O'Sullivan EJ, Johnson MR, et al. (1990) Increased incidence of lymphoproliferative disorder after immunosuppression with the monoclonal antibody OKT3 in cardiac-transplant recipients. N Engl J Med 323: 1723-1728.
  30. European best practice guidelines for renal transplantation (2002). Section IV: Long-term management of the transplant recipient. IV.6.1. Cancer risk after renal transplantation. Post-transplant lymphoproliferative disease (PTLD): prevention and treatment. Nephrol Dial Transplant 17: 31-33, 35-36.
  31. Morton M, Coupes B, Roberts SA, Johnson SL, Klapper PE, et al. (2014) Epstein–Barr Virus Infection in Adult Renal Transplant Recipients 14: 1619-1629.
  32. Martelius T, Lappalainen M, Palomäki M, Anttila VJ (2011) Clinical characteristics of patients with Epstein Barr virus in cerebrospinal fluid. BMC Infect Dis 11: 281.
  33. Aravindan AN, Moger V, Sakhuja V, Kohli HS, Varma N, et al. (2006) Hepatitis C virus related lymphoproliferative disorder in a renal transplant recipient. International Urology and Nephrology 38: 355-357.
  34. Imafuku A, Tanaka K, Marui Y, Ubara Y, Takaichi K, et al. (2018) Primary Central Nervous System Post-Transplant Lymphoproliferative Disorder Diagnosed by Peripheral Facial Nerve Palsy. Internal medicine 57: 1899-1903.
  35. Oliver M, Mitro G, Tenbrink P, Alharthi S, Bedford N, et al. (2019) No Increased Risk of Post-Transplant Lymphoproliferative Disorder Following Alemtuzumab Induction in Kidney Transplantation. Experimental and Clinical Transplantation 3: 320-325.
  36. Lim WH, Russ GR, Coates PT (2006) Review of Epstein-Barr virus and post-transplant lymphoproliferative disorder post-solid organ transplantation. Nephrology (Carlton) 11: 355-366.
  37. Nabors LB, Palmer CA, Julian BA, Przekwas AM, Kew CE (2009) Isolated central nervous system post-transplant lymphoproliferative disorder treated with high-dose intravenous methotrexate. Am J Transplant 9: 1243-1248.
  38. Yaginuma T, Yamamoto H, Mitome J, Tanno Y, Yamamoto I, et al. (2012) Successful treatment of monomorphic primary central nervous system post-transplantation lymphoproliferative disorder 5 years after kidney transplantation. Transpl Infect Dis 14: 102-106.
  39. Dotti G, Fiocchi R, Motta T, Gamba A, Gotti E, et al. (2000) Epstein-Barr virus-negative lymphoproliferate disorders in long-term survivors after heart, kidney, and liver transplant. Transplantation 69: 827-833.
  40. Twist CJ, Castillo RO (2013) Treatment of recurrent post-transplant lymphoproliferative disorder of the central nervous system with high-dose methotrexate. Case reports in transplantation 2013: 765230-765230.
  41. Krishnamurthy J, Greiner T, Bhatt V, White M, Bierman P (2014) LL-02: TEMOZOLOMIDE IN PRIMARY CENTRAL NERVOUS SYSTEM POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER (CNS-PTLD): A CASE SERIES. Neuro-Oncology 16: 119-119.
  42. Dugan JP, Haverkos BM, Villagomez L, Martin LK, Lustberg M, et al. (2018) Complete and Durable Responses in Primary Central Nervous System Posttransplant Lymphoproliferative Disorder with Zidovudine, Ganciclovir, Rituximab, and Dexamethasone. Clinical cancer research: an official journal of the American Association for Cancer Research 24: 3273-3281.
  43. Caillard S, Porcher R, Provot F, Dantal J, Choquet S, et al. (2013) Post-Transplantation Lymphoproliferative Disorder After Kidney Transplantation: Report of a Nationwide French Registry and the Development of a New Prognostic Score 31: 1302-1309.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Surgery

slot starlight princessslot gacor pgsoftakun gacor olympusrtp slot onlinejam gacor slot pg softtrik gacor slot aztecfitur scatter hitam slot mahjongsugar rush modal recehcheat apk engineslot mahjong gokil histerisinfo rtp harianrtp mahjong untungcheat mahjong bandar rungkatmodal receh olympusslot online thailandpola jitu starlightscatter naga hitamrtp gacor banjir wildslot88 jackpot kalitrik pola x5000olympus x500depo dana modal recehpg soft mudah gacorrahasia menang slotrtp balik modalcandu menang slot mahjongslot deposit danatips ampuh bermain slot mahjong waystrik slot sugar rushakun pro mahjong gacorrtp slot terjituslot mahjong ways gacorcara dapetin maxwin olympuspancing scatter mahjong ways 1rekomendasi slot mahjong ways 2scatter mahjong terbarupola mahjong ways hari inimahjong ways modal recehcuan mahjong waysdemo slot pg softnaga awal julyrtp slot awal julymahjong bulan mudamodal receh slotlink slot mahjongwinrate tinggi rtpslot server filipinavolatility pg softwaktu tepat slot gacorjam gacor saldo bancarfitur bonus lucky neko4 simulasi jackpot mahjongtrik sepuh mantan napiamantotorm1131