Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Review

Heart: first published as 10.1136/heartjnl-2020-318139 on 1 July 2021. Downloaded from http://heart.bmj.com/ on December 12, 2023 at Universidad Nacional Autonoma de Mexico.
Implications of cancer prior to and after
heart transplantation
Parvathi Mudigonda,1 Cecilia Berardi,1 Vishaka Chetram,2 Ana Barac,3
Richard Cheng ‍ ‍1

► Additional supplemental ABSTRACT outcomes in immunosuppressed patients with solid


material is published online Cancer and cardiovascular disease share many risk organ cancers.2 3 Accordingly, patients with active
only. To view, please visit the
journal online (http://d​ x.​doi.​ factors. Due to improved survival of patients with cancer, or recent cancer (non-­skin) have been traditionally
org/1​ 0.​1136/​heartjnl-­​2020-­​ the cohort of cancer survivors with heart failure referred excluded from transplant candidacy until they can
318139). for heart transplantation (HT) is growing. Specific be considered cured and/or the risk for long-­term
1
considerations include time interval between cancer recurrence is estimated as low. With recent advances
Department of Medicine,
Division of Cardiology,
treatment and HT, risk for recurrence and risk for de novo in cancer treatment, the prognosis of many cancers
University of Washington, malignancy (dnM). dnM is an important cause of post-­HT continues to rapidly improve, thus bringing into
Seattle, Washington, USA morbidity and mortality, with nearly a third diagnosed question the validity of historical survivorship
2
Department of Medicine, with malignancy by 10 years post-­HT. Compared with definitions and existing screening recommenda-
MedStar Washington Hospital
Center, Georgetown University,
the age-­matched general population, HT recipients have tions, and highlighting the potential need for new
Washington, DC, USA an approximately 2.5-­fold to 4-­fold increased risk of methods of risk stratification.
3
Department of Cardiology, developing cancer. HT recipients with prior malignancy In patients with a history of malignancy, different
MedStar Heart and Vascular show variable cancer recurrence rates, depending on durations of lapsed time may be optimal between
Institute, Georgetown years in remission before HT: 5% recurrence if >5 years completing cancer treatment and eligibility for
University, Washington, DC, USA
in remission, 26% recurrence if 1–5 years in remission HT. Among other factors, the interval depends on
Correspondence to and 63% recurrence if <1 year in remission. A myriad tumour type, response to cancer therapy, risk for
Dr Richard Cheng, Department of mechanisms influence oncogenesis following HT, recurrence and negative metastatic evaluation.3
of Medicine, Division of including reduced host immunosurveillance from While there are site-­specific recommendations for

Protected by copyright.
Cardiology, University of chronic immunosuppression, influence of oncogenic time from cancer treatment before listing for kidney
Washington Medical Center,
Seattle, WA 98195, USA;
viruses, and the cumulative intensity and duration of transplant, similar criteria have not been developed
​rkcheng@​uw.​edu immunosuppression. Conversely, protective factors or consistently adopted for HT. In the kidney trans-
include acyclovir prophylaxis, use of proliferation signal plant recommendations, time intervals are variable
Received 27 March 2021 inhibitors (PSI) and female gender. Management involves depending on the specific cancer type and stage,
Accepted 2 June 2021
Published Online First
reducing immunosuppression, incorporating a PSI ranging from 0 to 5 years.4 Conversely, for HT,
1 July 2021 for immunosuppression and heightened surveillance guidelines have suggested that cancer be in remis-
for allograft rejection. Cancer treatment, including sion for 5 years unless they are low grade.3
immunotherapy, may be cardiotoxic and lead to graft A review by Mistiaen5 compiled 13 retrospective
failure or rejection. Additionally, there exists a competing studies of 2017 paediatric and adult patients under-
risk to reduce immunosuppression to improve cancer going HT, finding that cancer recurrence inversely
outcomes, which may increase risk for rejection. A correlated with longer cancer-­free periods before
multidisciplinary cardio-­oncology team approach is transplant: 63% in those cancer-­free <1 year, 26%
recommended to optimise care and should include an in those cancer-­ free for 1–5 years and 5% with
oncologist, transplant cardiologist, transplant pharmacist, >5 years cancer-­free interval.5
palliative care, transplant coordinator and cardio-­ Table 1 (and online supplemental file 1)
oncologist. summarise the published studies that included
>20 HTr with history of PTM. Although
MALIGNANCY PRIOR TO HEART outcomes were different across studies, most
TRANSPLANTATION evaluated cancer-­ free survival, including the
Due to improved survival of patients with cancer and risk of recurrence, risk of new malignancy and
the use of cardiotoxic chemotherapeutic agents, an overall mortality. In a large analysis of 23 171
increasingly encountered scenario involves patients HTr through the Organ Procurement and Trans-
undergoing evaluation for cardiac transplantation plantation Network/United Network for Organ
with a history of pretransplant malignancy (PTM). Sharing (UNOS) database, history of malignancy
Registry data from the International Society for was associated with increased risk of cancer post-­
Heart and Lung Transplantation (ISHLT) showed HT. However, this risk was largely driven by post-­
that from 1992 to 2000, 3.7% of heart transplant transplant skin cancer.6 Another large study that
© Author(s) (or their
employer(s)) 2022. No recipients (HTr) had prior malignancy, which more used the UNOS database did not find a significant
commercial re-­use. See rights than doubled to 8.6% in 2010–2018.1 increase in 1-­year or 5-­year mortality in patients
and permissions. Published The rationale for cancer screening as part of the with PTM.7 In the subgroup matched cohort anal-
by BMJ. ysis, based on the PTM diagnosis, only haemato-
heart transplant (HT) evaluation is supported by
To cite: Mudigonda P, the concern for progression of previously indo- logical malignancies were associated with higher
Berardi C, Chetram V, et al. lent subclinical cancers with post-­HT immunosup- 1-­year post-­HT mortality, underscoring the impor-
Heart 2022;108:414–421. pression and the well-­ recognised risk of adverse tance of a cancer site-­specific approach.7
414   Mudigonda P, et al. Heart 2022;108:414–421. doi:10.1136/heartjnl-2020-318139
Review

Heart: first published as 10.1136/heartjnl-2020-318139 on 1 July 2021. Downloaded from http://heart.bmj.com/ on December 12, 2023 at Universidad Nacional Autonoma de Mexico.
Table 1 Selected studies investigating outcomes in patients with pretransplant malignancy
Study Study design, population Outcome Comments
Ladowski et al43 Retrospective, HTr 10-­year survival was 63% for PTM and 62% for no PTM Younger, leaner patients with a cured PTM show similar
(p=NS). long-­term survival to patients without PTM.
­ ivancos et al44
Fernández-V Retrospective, SOTR SOTR with PTM: pooled HR. PTM associated with increased risk of all-­cause
► All-­cause mortality: HR 1.51. mortality, cancer-­specific mortality and dnM
► Cancer-­related mortality: HR 3.13. development.
► dnM development: HR 1.92.
Sigurdardottir et al45 Retrospective, heart and lung Remission <1 year had worse survival than >1 year Cancer-­free survival of 5 years before heart or lung
recipients (p=0.044). transplant is associated with the lowest recurrence.
Beaty et al7 Retrospective, heart and lung PTM hazard of mortality in HTr, all cancer: PTM largely not associated with reduced survival
recipients ► 30-­day: HR 1.19, p=0.40. post-­HT, with notable exception for haematological
► 1 year: HR 0.87, p=0.35. malignancies.
► 5 years: HR 0.99, p=0.98.
PTM hazard of mortality in HTr, haematological:
► 30-­day: HR 1.82, p=0.04.
► 1 year: HR 1.93, p<0.001.
► 5 years: HR 1.54, p=0.01.
Mistiaen5 Systematic review, HTr Short-­term and long-­term post-­HT survival similar in PTM HT safe in PTM, especially with remission interval >5
vs no PTM. years.
Remission <1 year had markedly lower 5-­year survival Haematological malignancies pretransplant had worst
compared with a longer remission. post-t­ ransplant survival.
Yoosabai et al6 Retrospective, HTr PTM increased risk of post-­HT cancer. PTM increased the risk of post-­transplant malignancy,
► Any cancer: SHR 1.51. especially skin cancer.
► Skin: SHR 2.79. Older, white, men were risk factors for post-­transplant
► Solid organ malignancy: SHR 1.54. malignancy.
Delgado et al46 Retrospective, HTr ► 1-­year survival: PTM 95%, non-­PTM 93%. No significant differences of mortality incidence rates
► 5-­year survival: PTM 74%, non-­PTM 79%. or HR in HTr with PTM and without PTM. Mortality
► 10-­year survival: PTM 65%, no PTM 51% (p=0.048). declined in the recent time periods. Women had

Protected by copyright.
Cancer-­related mortality: leading cause of death in significantly better survival than men.
PTM (32.1%) and penultimate cause of death in no Overall PTM associated with higher post-­transplant
PTM (21.3%), after cardiovascular causes (22.8%). tumour incidence and haematological malignancies had
lower 10-­year survival compared with no PTM.
Please see online supplemental file 1 for the expanded version of this table.
dnM, de novo malignancy; HT, heart transplant; HTr, heart transplant recipients; PTM, pretransplant malignancy; SHR, subhazard ratio; SOTR, solid organ transplant recipient.

Recommendations for cancer screening pre-HT De novo malignancy (dnM) is defined as incident cancer occur-
Most clinical documents addressing cancer screening strate- ring after transplantation not directly related to the transplanted
gies have included age-­ appropriate and history-­ appropriate organ. Since 1995, a rising incidence of non-­lymphoma cancer
screening (figure 1), with addition of focused imaging (table 2). has been observed in HTr, largely attributable to an increase in
The ISHLT listing criteria acknowledges diversity of pre-­existing skin cancer.1 12 ISHLT registry data showed that between 1995
malignancies and outcomes in the setting of contemporary and 2018, 16% of HTr developed cancer within 5 years, 28%
oncology treatments,3 8 but specific recommendations by cancer at 10 years and more than 40% at 15 years after HT.1 HTr with
type are not provided. They recommend collaboration between malignancy have significantly lower survival compared with HTr
HT and oncology teams to assure individualised patient risk without malignancy or the general population.12 dnM was the
stratification. most common cause of death in the late-­transplant period, over-
taking graft failure at 5 years after HT.1
MALIGNANCY AFTER HT The most common malignancies in HTr are non-­ melanoma
Given the increased risk and high morbidity of cancer post-­HT, skin cancers (NMSC), solid malignancies, and rarely lymphomas
routine surveillance is recommended. The ISHLT guidelines (table 3 and online supplemental file 1). The ISHLT registry
recommend routine screening for breast, colon and prostate showed the incidence of skin cancer at 5 and 10 years after trans-
cancer in alignment with standard intervals used for the general plant was 9.6% and 18.5%, respectively. HTr with skin cancer
population. Additionally, HTr require close skin surveillance generally had a favourable prognosis and comparable survival with
with annual dermatological evaluations due to high risk for skin HTr without malignancy. The rates of developing a non-­cutaneous
cancers associated with chronic immunosuppression.9 solid malignancy at 5 and 10 years after transplant were 6.3% and
10.2%, respectively. Post-­transplant lymphoproliferative disease
De novo malignancy (PTLD) tended to be the most aggressive form of malignancy, with
Over the last three decades, the characteristics of HTr have the worst survival; however, the overall incidence was low, with
evolved, with a rise in higher risk individuals receiving HT. From 5-­year and 10-­year rates of 1% and 1.7%, respectively.1
1992 to 2000, 13.3% of HTr had diabetes mellitus; this more In summary, dnM is not only common in HTr but potentially
than doubled to 26.9% in 2010–2018, and HTr with a history of life-­limiting and a competing risk for death with allograft failure
dialysis increased from 3% to 4.8%.1 Due to steadily improving in the intermediate-­term to long-­term setting. Future research is
survival post-­HT, there are now HTr who are older, with chronic needed on whether systematic efforts on pretransplant selection
comorbidities, who will be exposed to prolonged immunosup- and post-­HT surveillance would reduce the risk associated with
pression, all well-­established risk factors for malignancy.10 11 dnM.
Mudigonda P, et al. Heart 2022;108:414–421. doi:10.1136/heartjnl-2020-318139 415
Review

Heart: first published as 10.1136/heartjnl-2020-318139 on 1 July 2021. Downloaded from http://heart.bmj.com/ on December 12, 2023 at Universidad Nacional Autonoma de Mexico.
Protected by copyright.
Figure 1 Monitoring for cancer pre-­HT and post-­HT. BRCA, breast cancer gene; EBV, Epstein-­Barr virus; HPV, human papilloma virus; HT, heart
transplantation; PTLD, post-­transplant lymphoproliferative disease.

Mechanisms of carcinogenesis common malignancies in the general population such as breast


Drivers of oncogenesis after transplantation are multidomain, and colorectal cancer are less commonly seen in HTr.12 15 16
with oncogenic viral infections, environmental exposures and Oncogenic viruses and malignancy associations in HTr include
a genetic predisposition all aggravated in the milieu of high-­ Epstein-­Barr virus (EBV) with lymphoma, human herpes virus
intensity and chronic immunosuppression. Long-­term immuno- 8 with Kaposi’s sarcoma, and human papilloma virus (HPV)
suppression, necessary to prevent rejection, is the single most with urogenital and anogenital cancers, cutaneous squamous cell
important risk factor for carcinogenesis in HTr. Parallels have carcinoma (SCC), and lip and oral cavity cancers.17–19
been drawn between solid organ transplant recipients (SOTRs) dnM development is closely related to the deliberate modula-
and immunocompromised individuals in that they share an tion of the innate immune system, with several proposed mech-
unusually high incidence of cancers uncommon in the general anisms: (1) reduced immunosurveillance of neoplastic cells and
population, including lymphomas, anogenital tumours and oncogenic viruses, allowing unchecked proliferation; (2) action
tumours related to oncogenic viruses.13 14 On the contrary, of oncogenic viruses from latent reactivation in the recipient,
acquired from the donor at the time of transplantation or after
transplantation; and (3) direct carcinogenic effects of immuno-
Table 2 Screening for cancer pre-­heart transplant suppressive medications.
Author Year Cancer screening recommendations
Mehra et al3
2006 ► Stool for occult gastrointestinal bleeding ×3.
Risk factors for dnM
► Colonoscopy if indicated or >50 years of age. Various risk factors for dnM were identified from several large
► Mammography if indicated or women >40 years of age. cohort analyses of HTr and other SOTRs. Older age at the time
► Gynaecological examination/Pap test in sexually active of transplant and male gender were consistently observed. Other
women >18 years of age. strong risk factors were chronic immunosuppression, influence
► PSA and digital rectal examination in men >50 years of oncogenic viruses, retransplantation and malignancy prior to
of age.
HT. Less robust risk factors include recipient smoking history,
► Serum protein electrophoresis/urine protein
electrophoresis if multiple myeloma suspected. use of induction immunosuppression, radiation exposure prior
Mehra et al8 2016 In addition to the above, consider the following:
to transplantation and genetic variance (figure 2).11–13 15 18 19
► Chest X-­ray and chest CT. A study using the ISHLT registry between 2000 and 2011
► Oesophagogastroduodenoscopy. identified several risk factors for developing cutaneous malig-
► Colonoscopy. nancy, non-­cutaneous malignancy and PTLD within 5 years of
Pap, Papanicolaou smear test; PSA, prostate-­specific antigen. HT.12 In cutaneous malignancies, the strongest association was
416 Mudigonda P, et al. Heart 2022;108:414–421. doi:10.1136/heartjnl-2020-318139
Review

Heart: first published as 10.1136/heartjnl-2020-318139 on 1 July 2021. Downloaded from http://heart.bmj.com/ on December 12, 2023 at Universidad Nacional Autonoma de Mexico.
Table 3 Selected studies investigating de novo malignancy in heart transplant recipients
Study Conclusions
Crespo-­Leiro et al,13 retrospective ► Most dnMs were skin cancers (50%) and NHL (10%).
► ATG use was a strong risk factor for lymphomas and non-­skin cancers in the absence of acyclovir prophylaxis.
► Male gender and age ≥45 years were risk factors for all cancers, except lymphoma.
► Smoking before HT was a risk factor for all cancers, except skin cancer and lymphoma.
Kellerman et al,47 prospective ► Oncogenic viruses had a strong association with dnM: EBV with lymphoma and HPV with cervical cancer.
► Older age and retransplantation were significant risk factors for dnM.
Collett et al,48 retrospective ► Cancer incidence in HTr greater than twofold compared with general population.
► NMSC was the most common.
► Low incidence of breast and cervical cancer in SOTRs, overall.
► Male gender and older age were common in dnM after HT.
Jiang et al,49 retrospective ► NMSC was excluded (lower SIR in this study).
► Most common cancers in HTr were lymphomas, oral cancers and lung cancer.
► Lymphomas most common in the first year after HT and the most common cancer in young HTr.
► Risk factors for malignancy development in HTr were smoking, sun exposure and prior malignancy.
Engels et al,14 retrospective ► HTr at an increased risk of developing dnM compared with the general population.
Sampaio et al,11 retrospective ► Cancer incidence higher in thoracic organ recipients compared with abdominal organ recipients.
► In all SOTRs, older age and male gender were risk factors for malignancy.
► Most common dnM in HTr were lung cancer, PTLD and colorectal cancer.
Higgins et al,15 retrospective ► NMSC was excluded (declining malignancy risk over 15 years and lower malignancy rates).
► No significant gender differences were observed.
► Lung cancer, lymphomas and melanomas were the most common cancers.
► Risk factors for dnM were older age and earlier era of transplant.
► 5-­year survival: lung cancer 21%, lymphoma 32%, prostate cancer and melanoma >50%.

Youn et al,12 retrospective ► Between 1 and 5 years after HT, >10% of HTr developed dnM.
► HTr with dnM had worse survival than HTr without dnM.
► Overall cancer incidence rising during the studied periods, largely due to skin cancer incidence.

Protected by copyright.
Jäämaa-­Holmberg et al, 50 prospective ► Compared with general population, HTr had higher cancer incidence, with greater morbidity and three times the mortality
rate.
► Most common cancers were NMSC (BCC 31% and SCC 21%), followed by NHL (13.5%).
► DnM risk increased temporally over the study duration.
► Male gender was a powerful risk factor, with over 95% of all observed cancers in men.
Please see online supplemental file 1 for the expanded version of this table.
ATG, antithymocyte globulin; BCC, basal cell carcinoma; dnM, de novo malignancy; EBV, Epstein-­Barr virus; HPV, human papilloma virus; HT, heart transplant; HTr, heart
transplant recipient; NHL, non-­Hodgkin's lymphoma; NMSC, non-­melanoma skin cancer; PTLD, post-­transplant lymphoproliferative disorder; SCC, squamous cell carcinoma; SIR,
standardised incidence ratio; SOTR, solid organ transplant recipient.

Figure 2 Risk factors for malignancy in the general population and specific to heart transplant (figure made with Biorender.com). ATG,
antithymocyte globulin; EBV, Epstein-­Barr virus; HPV, human papilloma virus; UV, ultraviolet.
Mudigonda P, et al. Heart 2022;108:414–421. doi:10.1136/heartjnl-2020-318139 417
Review

Heart: first published as 10.1136/heartjnl-2020-318139 on 1 July 2021. Downloaded from http://heart.bmj.com/ on December 12, 2023 at Universidad Nacional Autonoma de Mexico.
with older age, later calendar year of transplant, congenital heart lymphoma.17 Serological EBV mismatch at the time of transplant
disease and retransplant/graft failure. Other risk factors included (EBV-­positive donor to an EBV-­negative recipient) results in a
azathioprine (AZA) use compared with mycophenolate mofetil 12-­fold increase in the risk for PTLD.29 Nearly 50%–80% of
(MMF), cytomegalovirus mismatch, taller stature, use of inter- PTLD cases are associated with EBV, especially in the first few
leukin-2 receptor (IL-­2R) antagonist or muromonab-­CD3 induc- years after transplantation. T cell immunity is weakened by cyto-
tion, and readmission within 1 year of HT.12 lytic immunosuppressive agents in transplant recipients, leading
Risk factors for non-­cutaneous solid cancer were older age, to impaired surveillance against EBV and other oncoviruses.29
recipient former tobacco use, later calendar year of transplant The incidence of PTLD in HTr is between 1.2% and 6.5%,30
and readmission within 1 year of HT. Risk factors for developing with a median time to diagnosis of 3.3 years.31 Increased risk of
PTLD were cell cycle inhibitor non-­use or AZA as compared developing PTLD is driven by age extremes (<18 years and >50
with MMF use, antithymocyte globulin induction, recipient EBV years), use of tacrolimus, use of AZA, and most significantly with
seronegativity, readmission within 1 year of HT and obesity.12 lymphocyte-­depleting antibody agents used for either induction
or rejection treatment. The risk was reduced when antiviral
prophylaxis was administered with lymphocyte-­depleting anti-
Immunosuppression
bodies, particularly with acyclovir.13 29 Overall, the cumulative
Whether induction therapy is directly associated with dnM after
degree and duration of immunosuppression drives the risk for
HT remains unclear. While the above studies suggested an asso-
PTLD, rather than exposure to any single specific medication.
ciation,12 13 other studies found similar risk for malignancy with
Survival was profoundly worse after developing PTLD, with
induction and non-­induction regimens.20 21
only a third of HTr alive at 5 years.31 32 However, survival
Direct carcinogenic effects of chronic immunosuppressive
improved significantly if complete remission was achieved within
medications are well recognised, particularly with AZA and
3 months and subsequent rate of relapse was low at 13%. Use
calcineurin inhibitors (CNI). For example, in a cohort that
of a rituximab-­based treatment regimen compared with other
included any SOTR, AZA was associated with a 56% greater
chemotherapy for treating PTLD was associated with improved
risk of developing cutaneous SCC compared with other immu-
survival in multiple cohorts.31 33 In all cases, the basis for PTLD
nosuppressants.22 23 MMF is a non-­ competitive inhibitor
treatment begins with dose reduction of immunosuppression to
involved in purine synthesis. A 2005 multicentre randomised
restore host immunity.
trial comparing MMF with AZA over 3 years showed improved
survival in patients on MMF. dnM occurred in 15.6% of the
AZA group and 12.5% of the MMF group; however, this did not Skin cancer

Protected by copyright.
reach statistical significance.24 These findings were supported in Skin cancers are the most common post-­transplant malignancy
a 2006 ISHLT registry analysis of HTr between 1995 and 1997 and account for 40%–50% of total cancers.34 By 15 years post-­
that showed a lower incidence of dnM in immunosuppressive HT, nearly a third of HTr will have skin cancer of any type.1
regimens with MMF as compared with AZA.25 SCC and basal cell carcinoma (BCC) comprise >90% of all skin
Sirolimus (SRL) and everolimus are proliferation signal inhib- cancers.35
itors (PSI) belonging to the mammalian target of rapamycin Risk factors for skin cancer include ultraviolet radiation expo-
(mTOR) inhibitor family. The mTOR pathway regulates cell sure, older age at transplant, male gender, white race, smoking,
growth and survival and is frequently dysregulated in various high-­intensity immunosuppression, HPV infection, human
malignancies.26 Hence, it would be anticipated that SRL might leucocyte antigen DR (HLA-­DR) mismatches and a history of
reduce the risk of cancer. Supporting this, studies have demon- NMSC.12 34 36 Immunosuppression regimens with AZA and/or
strated a reduction in risk of malignancy and NMSC in post-­ cyclosporine had a higher incidence of SCC development.27 34
transplant patients treated with SRL.27 The lower incidence of MMF use was associated with a lower risk of SCC and a greater
malignancy following conversion from CNI to mTOR inhibitors risk of BCC, compared with AZA.34 PSI reduced the risk of
has been attributed to antiproliferative effects, particularly from NMSC.27
suppressing tumour growth and angiogenesis.27 Despite the high incidence of skin cancer and risk for metas-
A recent study of 523 patients who underwent HT between tasis, survival is generally favourable and only slightly worse
1994 and 2016 examined whether conversion from a CNI-­ than HTr without malignancy.1 12 36 This may be related to the
based to SRL-­based regimen was associated with decreased risk heightened dermatological surveillance in HTr, leading to early
of malignancy. Over a median follow-­up of 10 years post-­HT, detection of skin cancers.
the risk for malignancies (excluding NMSC) was 13% in those
on SRL and 31% in those on CNI-­based regimens. Findings CANCER MANAGEMENT
were consistent for all dnMs and for PTLD.28 In this study, late Data are sparse on specific cancer treatment regimens in HTr.
survival was better in those on SRL-­based regimens who were Prevailing considerations include (1) the need for clinical phar-
free from malignancy. macists to review medications for potential drug–drug inter-
While prospective trials of PSI use in HT specifically to reduce actions particularly with CNIs and mTOR inhibitors; (2) the
cancer risk are lacking, these data suggest that individuals at high desire to minimise immunosuppression during cancer treatment;
risk for malignancy after HT (such as those with prior cancer) (3) potential antioncogenic effects of mTOR inhibitors; and
should be considered for a PSI-­based regimen. (4) heightened concern for cardiotoxicity risk in a vulnerable
cohort. Discussion on treatment for each cancer type is beyond
Post-transplant lymphoproliferative disease the scope of this review. The primary focus will be on immuno-
PTLD describes a heterogeneous group of lymphoid disorders therapy as there are unique considerations for the HTr.
that result from abnormal lymphocyte proliferation in the setting
of chronic immunosuppression. Disease severity is highly vari- Immunotherapy
able and ranges from reactive lymphoid hyperplasia to aggres- In recent years, modulation of the immune system, specifically T
sive malignant lymphomas, most commonly diffuse large B cell cell activity, has become a new and important aspect of targeted
418 Mudigonda P, et al. Heart 2022;108:414–421. doi:10.1136/heartjnl-2020-318139
Review

Heart: first published as 10.1136/heartjnl-2020-318139 on 1 July 2021. Downloaded from http://heart.bmj.com/ on December 12, 2023 at Universidad Nacional Autonoma de Mexico.
population characteristics and the lack of standardised diag-
nostic criteria.37 38
Little is known regarding ICI use in HTr as they have been
generally excluded from clinical trials. In a retrospective case
series, three of four HTr treated with ICI had biopsy-­proven
allograft rejection with cardiac dysfunction, including one
death.39 The fourth patient, who received pembrolizumab for
metastatic melanoma and remained on tacrolimus monotherapy,
was alive at 15 months of follow-­up; no specific demographic,
malignancy-­ related or graft-­
related differences were observed
that explained this favourable outcome. A systematic review of
83 patients with kidney, liver or heart transplants treated with
ICI showed a 39.8% incidence of allograft rejection, with the
vast majority within the first 2 weeks of treatment.40 Overall, 48
(57.8%) patients died. Of six HT patients, only one had rejec-
tion, but four out of six died, suggesting that factors beyond
rejection may have a prognostic role. Recently, a pharmacovigi-
lance analysis identified 96 reports of any organ transplant rejec-
tion after ICI exposure, including five HTr. The median time
to rejection in HTr was 5 days with 20% short-­term mortality.
Across all SOTRs, anti-­programmed death-­1 (PD-­1) and anti-­
Figure 3 Specific considerations for immunotherapy in heart
programmed death-­ ligand 1 (PD-­ L1) were more frequently
transplantation recipients. BiTEs, bispecific T cell engagers; CAR-­T,
implicated than cytotoxic T-­ lymphocyte-­associated protein 4
chimeric antigen receptor T cells; ICIs, immune checkpoint inhibitors.
(CTLA-­4) .41 Unfortunately, the sample sizes in these studies are
too small to inform optimal use of ICI in HTr. Due to the rare
cancer therapy. Immune checkpoint inhibitors (ICIs) were nature of ICI use in HTr, multicentre registries were developed,
the first class to be used in clinical practice; chimeric antigen including a thoracic-­focused registry at the University of Utah39
receptor T cells (CAR-­T) therapy and bispecific T cell engagers and a SOTR registry at the University of Washington.

Protected by copyright.
(BiTEs) followed in recent years. CAR-­ T and BiTEs use single-­ chain variable fragments to
ICIs are monoclonal antibodies that enhance T cell prolifer- direct the activity of cytotoxic T lymphocytes against antigens
ation and activation by removing inhibitory feedback through expressed by tumour cells. The most severe adverse effect is
different pathways. ICI-­ associated cardiovascular toxicity can cytokine release syndrome. Generally observed early in the treat-
present with myocarditis, non-­ inflammatory cardiomyopathy, ment course, this severe inflammatory state can lead to haemo-
arrhythmia, pericardial disease or vasculitis. In the general dynamic instability, arrhythmia, tachycardia and fluid overload.
population, the incidence of cardiovascular adverse effects in Data for these newer therapies in HT are lacking.
patients treated with ICIs has been reported to be as low as 0% While immunotherapy is increasingly used for cancer treat-
and as high as 5.5%, likely reflecting differences in methods and ment, its mechanism of action poses specific risks to the HT

Figure 4 Multidisciplinary approach to cancer treatment in heart transplantation recipients.


Mudigonda P, et al. Heart 2022;108:414–421. doi:10.1136/heartjnl-2020-318139 419
Review

Heart: first published as 10.1136/heartjnl-2020-318139 on 1 July 2021. Downloaded from http://heart.bmj.com/ on December 12, 2023 at Universidad Nacional Autonoma de Mexico.
Table 4 Gaps in knowledge and future areas of research
Scenario Current practice Gaps in knowledge and implications
Malignancy prior to HT:
► What screening should be required prior to HT? ► Standard population screening and incorporation of ► For patients in shock, can some aspects of this
► What is the optimal time interval after cancer imaging such as CT chest and abdomen. be skipped? In particular, colonoscopy may be
treatment prior to HT? ► Current recommendations and most programmes wait challenging in an unstable patient.
► How do you estimate risk for recurrence or secondary 5 years after cancer prior to HT. ► Survival and risk for recurrence varies by cancer type,
malignancy in those with prior cancer prior to HT? ► Current risk estimates use standard oncological stage and treatment. With improving treatment for
risk tools and do not account for chronic cancer, can HT be considered sooner in those with
immunosuppression after HT. good prognosis?
► The impact of immunosuppression on risk for
recurrence likely varies by cancer type. Additional
studies are needed on this topic.
Risk for cancer after HT:
► What is the optimal screening after HT? ► Despite increased risk for cancer, standard population ► It remains unclear whether intensified screening for
► Should immunosuppression regimens be tailored screening with addition of skin examinations are cancer post-­HT would lead to earlier detection or
based on risk for cancer? performed. better long-­term outcomes. There is a need to balance
► Should immunosuppression be minimised as much ► Most HT programmes use standard protocols and do additional testing and risk for incidental findings with
as possible? not tailor regimens based on risk. benefit.
► Most HT programmes have set dosing for ► Rather than a reactive approach, data are needed on
immunosuppression that is reduced over time and whether a pre-­emptive approach may improve long-­
then reduced further when there is a complication. term outcomes in high-­risk patients. Considerations
include avoiding induction immunosuppression or an
early transition to sirolimus.
► The optimal immunosuppression regimen prevents
rejection, and coronary vasculopathy while minimising
risk for malignancy needs further study.
Management of post-­HT cancer:
► How much can immunosuppression be reduced in ► Most programmes reduce immunosuppression in ► Can HT patients be reduced to monotherapy or even
those with active cancer? those with active cancer. temporarily hold immunosuppression during cancer

Protected by copyright.
► Is there increased risk for cardiotoxicity in HT ► Risk estimates for cardiotoxicity from the general treatment?
patients? population are used in HT. ► Are HT patients more susceptible to cardiotoxicity
► Is immunotherapy an option in HT patients? ► There are limited data on immunotherapy in HT from cancer treatments such as anthracyclines,
patients. tyrosine kinase inhibitors or radiation therapy?
► The use of ICI will continue to increase, creating a
difficult decision-­making scenario for HT transplants
with cancer. Research is paramount to understanding
whether ICI therapy is a viable option in HTr,
which patients are at increased risk, what type of
surveillance is necessary and how to manage HTr who
experience ICI-­associated rejection.
HT, heart transplantation; HTr, heart transplant recipients; ICI, immune checkpoint inhibitor.

population (figure 3). A paucity of data is available to estimate directions for research. Key considerations are summarised in
the risk of rejection or how best to modulate immunosuppres- table 4.
sion regimens during immunotherapy. Both rejection and the
malignancies treated with immunotherapy carry high mortality,
CONCLUSION
making the risk–benefit balance challenging.
With progressive improvements in cancer treatment, there is
a growing cohort of long-­term cancer survivors that develop
MULTIDISCIPLINARY APPROACH FOR INDIVIDUALISED significant cardiovascular toxicities including severe HF. Multi-
CARDIO-ONCOLOGY CARE disciplinary consideration is necessary for decision-­ making
Given the complex decision-­making, a multidisciplinary cardio-­ to proceed with HT based on risk for recurrence, mitigating
oncology approach is recommended. This should ideally include risk for dnM by tailoring immunosuppression and the need to
the HT cardiology team, an oncologist, pharmacy, palliative care balance immunosuppression with cancer treatment in those with
and a cardio-­oncologist. These models are increasingly common dnM. Given the rapid incorporation of immunotherapy into
in other cohorts and should be adopted for the HTr that many cancer treatment regimens, specific implications for HTr
develops cancer (figure 4). Additionally, given the large mental need to be recognised. These gaps in knowledge require further
and psychosocial challenges placed on the patient, early involve- exploration to develop an optimal holistic treatment framework
ment of palliative care can assist with decision-­making and care for a cohort that will be increasingly encountered.
planning in the setting of an uncertain prognostic trajectory.42
Twitter Richard Cheng @RichardKCheng2
FUTURE AREAS OF RESEARCH Contributors All authors have read and approved the manuscript. All authors
An international standardised collaboration for studying this contributed significantly to the final manuscript with design of the review, drafting of
unique population is currently lacking but is necessary to achieve the sections, revisions and critical review prior to submission.
a sufficiently sized cohort through a common protocol. Given the Funding The authors have not declared a specific grant for this research from any
rapid improvements in cancer treatment, this field will continue funding agency in the public, commercial or not-­for-­profit sectors.
to evolve and there remain many gaps in knowledge and future Competing interests None declared.

420 Mudigonda P, et al. Heart 2022;108:414–421. doi:10.1136/heartjnl-2020-318139


Review

Heart: first published as 10.1136/heartjnl-2020-318139 on 1 July 2021. Downloaded from http://heart.bmj.com/ on December 12, 2023 at Universidad Nacional Autonoma de Mexico.
Patient consent for publication Not required. 23 Cho HG, Kuo KY, Xiao K, et al. Azathioprine and risk of multiple keratinocyte cancers. J
Am Acad Dermatol 2018;78:27–8.
Provenance and peer review Commissioned; externally peer reviewed.
24 Eisen HJ, Kobashigawa J, Keogh A, et al. Three-­year results of a randomized, double-­
Supplemental material This content has been supplied by the author(s). It blind, controlled trial of mycophenolate mofetil versus azathioprine in cardiac
has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have transplant recipients. J Heart Lung Transplant 2005;24:517–25.
been peer-­reviewed. Any opinions or recommendations discussed are solely those 25 O’Neill JO, Edwards LB, Taylor DO. Mycophenolate mofetil and risk of developing
of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and malignancy after orthotopic heart transplantation: analysis of the transplant registry
responsibility arising from any reliance placed on the content. Where the content of the International Society for Heart and Lung Transplantation. J Heart Lung
includes any translated material, BMJ does not warrant the accuracy and reliability Transplant 2006;25:1186–91.
of the translations (including but not limited to local regulations, clinical guidelines, 26 Guba M, von Breitenbuch P, Steinbauer M, et al. Rapamycin inhibits primary and
terminology, drug names and drug dosages), and is not responsible for any error metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial
and/or omissions arising from translation and adaptation or otherwise. growth factor. Nat Med 2002;8:128–35.
27 Cangemi M, Montico B, Faè DA, et al. Dissecting the Multiplicity of Immune Effects of
ORCID iD Immunosuppressive Drugs to Better Predict the Risk of de novo Malignancies in Solid
Richard Cheng http://orcid.org/0000-0001-5994-5998 Organ Transplant Patients. Front Oncol 2019;9:160.
28 Asleh R, Clavell AL, Pereira NL, et al. Incidence of Malignancies in Patients Treated
REFERENCES With Sirolimus Following Heart Transplantation. J Am Coll Cardiol 2019;73:2676–88.
1 Khush KK, Cherikh WS, Chambers DC, et al. The International Thoracic Organ 29 Dierickx D, Habermann TM. Post-­Transplantation lymphoproliferative disorders in
Transplant Registry of the International Society for Heart and Lung Transplantation: adults. N Engl J Med 2018;378:549–62.
Thirty-­sixth adult heart transplantation report - 2019; focus theme: Donor and 30 Nagle SJ, Reshef R, Tsai DE. Posttransplant lymphoproliferative disorder in solid organ
recipient size match. J Heart Lung Transplant 2019;38:1056–66. and hematopoietic stem cell transplantation. Clin Chest Med 2017;38:771–83.
2 Conraads VM, Denollet J, Vorlat A, et al. Screening for solid organ malignancies prior 31 Kumarasinghe G, Lavee O, Parker A, et al. Post-­Transplant lymphoproliferative disease
to heart transplantation. Transplantation 2001;71:1481–3. in heart and lung transplantation: defining risk and prognostic factors. J Heart Lung
3 Mehra MR, Kobashigawa J, Starling R, et al. Listing criteria for heart transplantation: Transplant 2015;34:1406–14.
International Society for Heart and Lung Transplantation guidelines for the care of 32 Opelz G, Döhler B. Lymphomas after solid organ transplantation: a collaborative
cardiac transplant candidates--2006. J Heart Lung Transplant 2006;25:1024–42. transplant study report. Am J Transplant 2004;4:222–30.
4 Batabyal P, Chapman JR, Wong G, et al. Clinical practice guidelines on wait-­listing for 33 Evens AM, David KA, Helenowski I, et al. Multicenter analysis of 80 solid
kidney transplantation: consistent and equitable? Transplantation 2012;94:703–13. organ transplantation recipients with post-­transplantation lymphoproliferative
5 Mistiaen WP. Heart transplantation in patients with previous malignancy. An overview. disease: outcomes and prognostic factors in the modern era. J Clin Oncol
Acta Cardiol 2015;70:123–30. 2010;28:1038–46.
6 Yoosabai A, Mehta A, Kang W, et al. Pretransplant malignancy as a risk factor 34 Mittal A, Colegio OR. Skin cancers in organ transplant recipients. Am J Transplant
for posttransplant malignancy after heart transplantation. Transplantation 2017;17:2509–30.
2015;99:345–50. 35 Fortina AB, Caforio AL, Piaserico S, et al. Skin cancer in heart transplant recipients:
7 Beaty CA, George TJ, Kilic A, et al. Pre-­transplant malignancy: an analysis of outcomes frequency and risk factor analysis. J Heart Lung Transplant 2000;19:249–55.

Protected by copyright.
after thoracic organ transplantation. J Heart Lung Transplant 2013;32:202–11. 36 Brewer JD, Colegio OR, Phillips PK, et al. Incidence of and risk factors for skin cancer
8 Mehra MR, Canter CE, Hannan MM, et al. The 2016 International Society for Heart after heart transplant. Arch Dermatol 2009;145:1391–6.
Lung Transplantation listing criteria for heart transplantation: a 10-­year update. J 37 Ball S, Ghosh RK, Wongsaengsak S, et al. Cardiovascular Toxicities of
Heart Lung Transplant 2016;35:1–23. Immune Checkpoint Inhibitors: JACC Review Topic of the Week. J Am Coll Cardiol
9 Costanzo MR, Dipchand A, Starling R, et al. The International Society of Heart and 2019;74:1714–27.
Lung Transplantation guidelines for the care of heart transplant recipients. J Heart 38 Lyon AR, Yousaf N, Battisti NML, et al. Immune checkpoint inhibitors and
Lung Transplant 2010;29:914–56. cardiovascular toxicity. Lancet Oncol 2018;19:e447–58.
10 Miao Y, Everly JJ, Gross TG, et al. De novo cancers arising in organ transplant 39 Daud A, Mehra MR, Siu A, et al. Immune checkpoint inhibitors in heart or lung
recipients are associated with adverse outcomes compared with the general transplantation: early results from a registry initiative. J Heart Lung Transplant
population. Transplantation 2009;87:1347–59. 2020;39:604–6.
11 Sampaio MS, Cho YW, Qazi Y, et al. Posttransplant malignancies in solid organ adult 40 d’Izarny-­Gargas T, Durrbach A, Zaidan M. Efficacy and tolerance of immune
recipients: an analysis of the U.S. national transplant database. Transplantation checkpoint inhibitors in transplant patients with cancer: a systematic review. Am J
2012;94:990–8. Transplant 2020;20:2457–65.
12 Youn J-­C, Stehlik J, Wilk AR, et al. Temporal trends of de novo malignancy 41 Nguyen LS, Ortuno S, Lebrun-­Vignes B, et al. Transplant rejections associated with
development after heart transplantation. J Am Coll Cardiol 2018;71:40–9. immune checkpoint inhibitors: a pharmacovigilance study and systematic literature
13 Crespo-­Leiro MG, Alonso-­Pulpón L, Vázquez de Prada JA, et al. Malignancy after review. Eur J Cancer 2021;148:36–47.
heart transplantation: incidence, prognosis and risk factors. Am J Transplant 42 Cheng R, Kirkpatrick J, MSorror M. Cardio-­Oncology and the intersection of
2008;8:1031–9. cancer and cardiotoxicity: the role of palliative care. J Am Coll Cardiol CardioOnc
14 Engels EA, Pfeiffer RM, Fraumeni JF, et al. Spectrum of cancer risk among US solid 2019;2:314–7.
organ transplant recipients. JAMA 2011;306:1891–901. 43 Ladowski SD, Abel M, Beatty L, et al. Long-­term follow-­up of hearttransplant
15 Higgins RS, Brown RN, Chang PP, et al. A multi-­institutional study of malignancies recipients with pre-­transplant malignancies. Tex Heart Inst J 2006;33:27–30.
after heart transplantation and a comparison with the general United States 44 Fernández-­Vivancos C, Paniagua-­Martín MJ, Marzoa-­Rivas R, et al. Long-­term
population. J Heart Lung Transplant 2014;33:478–85. outcome in heart transplant patients with pretransplant malignancies. Transplant Proc
16 Wong G, Au E, Badve SV, et al. Breast cancer and transplantation. Am J Transplant 2010;42:3006–10.
2017;17:2243–53. 45 Sigurdardottir V, Bjortuft O, Eiskjær H, et al. Long-­term follow-­up of lung and heart
17 Dharnidharka VR, Webster AC, Martinez OM, et al. Post-­transplant lymphoproliferative transplant recipients with pre-­transplant malignancies. J Heart Lung Transplant
disorders. Nat Rev Dis Primers 2016;2:15088. 2012;31:1276–80.
18 Geissler EK. Post-­transplantation malignancies: here today, gone tomorrow? Nat Rev 46 Delgado JF, Alonso-­Pulpón L, Mirabet S, et al. Cancer incidence in heart transplant
Clin Oncol 2015;12:705–17. recipients with previous neoplasia history. Am J Transplant 2016;16:1569–78.
19 Buell JF, Gross TG, Woodle ES. Malignancy after transplantation. Transplantation 47 Kellerman L, Neugut A, Burke B, et al. Comparison of the incidence of de novo solid
2005;80:S254–64. malignancies after heart transplantation to that in the general population. Am J
20 Briasoulis A, Inampudi C, Pala M, et al. Induction immunosuppressive therapy in Cardiol 2009;103:562–6.
cardiac transplantation: a systematic review and meta-­analysis. Heart Fail Rev 48 Collett D, Mumford L, Banner NR, et al. Comparison of the incidence of malignancy
2018;23:641–9. in recipients of different types of organ: a UK registry audit. Am J Transplant
21 Hertig A, Zuckermann A. Rabbit antithymocyte globulin induction and risk of 2010;10:1889–96.
post-­transplant lymphoproliferative disease in adult and pediatric solid organ 49 Jiang Y, Villeneuve PJ, Wielgosz A, et al. The incidence of cancer in a population-­based
transplantation: an update. Transpl Immunol 2015;32:179–87. cohort of Canadian heart transplant recipients. Am J Transplant 2010;10:637–45.
22 Jiyad Z, Olsen CM, Burke MT, et al. Azathioprine and risk of skin cancer in organ 50 Jäämaa-­Holmberg S, Salmela B, Lemström K, et al. Cancer incidence and mortality
transplant recipients: systematic review and meta-­analysis. Am J Transplant after heart transplantation - A population-­based national cohort study. Acta Oncol
2016;16:3490–503. 2019;58:859–63.

Mudigonda P, et al. Heart 2022;108:414–421. doi:10.1136/heartjnl-2020-318139 421

You might also like