Abstract
Use of immune checkpoint inhibitors (ICIs), a revolutionary treatment in modern oncology, is frequently complicated by immune-related adverse events (irAEs), which can be confused with infections, and vice versa, thus complicating management decisions. In this study, we review the published cases of infections as simulators of irAEs in cancer patients.
Keywords: immune checkpoint inhibitors, infection, toxicity
The discovery that malignant cells can prevent recognition and destruction by the immune system through the expression of immunoregulatory molecules, such as cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and the PD-1 ligand (PD-L1), has dramatically changed the landscape of medical oncology. Thus, the development of immune checkpoint inhibitors (ICIs), which are monoclonal antibodies that inhibit the aforementioned molecules, has revolutionized cancer care [1]. Unlike conventional chemotherapy that kills malignant cells, ICIs work by inhibiting the pathways cancer cells use to evade host immune recognition.
After the implementation of ICIs in cancer treatment, a plethora of autoimmune phenomena known as immune-related adverse events (irAEs) presented as a consequence of the immune checkpoint (ICP) blockade, with reported incidences ranging from 54% to 76% [2]. Immune-related adverse events may affect any system of the body and, occasionally, can simulate a variety of infections such as colitis, encephalitis, or pneumonitis [2]. Management of moderate to severe irAEs often involves therapy with glucocorticoids and other immunosuppressants, such as antitumor necrosis factor agents, in case of corticosteroid refractoriness. More importantly, this immunosuppressive therapy can be complicated by opportunistic infections [3]. In contrast, little is known regarding infections as simulators of irAEs, in the absence of immunosuppressive therapy. These infections may appear coincidentally or be unmasked by a dysregulated inflammatory immune response due to ICIs. In this sense, they have been recently cataloged as “infections due to dysregulated immunity (ITI-ID)” [4]. This hypothesis is based on several reports of infections during ICI treatment without additional immunosuppression and builds on the fact that patients with hereditary CTLA-4 dysfunction or mutations are more prone to present with recurrent infections, particularly respiratory infections, including tuberculosis [5].
Identifying infections that simulate irAEs is extremely important because the management is completely different. Misdiagnosing infections can lead to delayed diagnosis and treatment and a deterioration of the infectious condition due to the treatment with corticosteroids and other immunosuppressants used for the management of the suspected irAEs. Finally, diagnosis of irAEs requires the exclusion of other infectious or inflammatory causes.
In this study, we sought to review infections simulating irAEs. We do not discuss the unmasking of indolent or latent infections by ICIs that simulate progression of underlying cancer (eg, exacerbation of preexisting mycobacterial or fungal lung infection simulating lung cancer progression) (Supplementary Material References 1–11).
METHODS
Search Strategy and Selection Criteria
A comprehensive search of the literature was constructed and performed by a qualified medical librarian (R.S.H.). Medline (Ovid), Embase (Ovid), Scopus, Cochrane, and Google Scholar were queried, from database inception until January 2022, using both controlled vocabulary and natural language terms for ICIs and viral, bacterial, or fungal infections (see Supplementary Materials). A total of 291 articles and abstracts were retrieved with the search strategy, and 22 were finally included in the manuscript.
RESULTS
Infections Simulating Toxicities
A variety of viral, bacterial and fungal infections have been sporadically described as simulators of irAEs in cancer patients treated with various ICIs with no additional immunosuppression (Table 1, Supplementary Table 1).
Table 1.
Organ/System Involved | Pathogen | Syndrome | Implicated ICI (Time From ICI Start) | Underlying Tumor | Reference |
---|---|---|---|---|---|
CNS | VZV | Encephalitis | Nivolumab (12 cycles) | Metastatic lung adenocarcinoma | Watanabe et al [9] |
VZV | Vasculopathy | Nivolumab (NA) | Lung | Ursu et al [10] | |
VZV | Ramsay-Hunt syndrome + ataxic neuropathy | Nivolumab (13 cycles) | NSCLC | Sakoh et al [11] | |
EBV | Cerebellar ataxia | Pembrolizumab (3 cycles) | Lung adenocarcinoma | Saikawa et al [13] | |
Lung | CMV | … | … | … | … |
Pneumocystis jirovecci | Pneumonitis | Nivolumab (3 cycles) | NSCLC | Liu et al [3] | |
Aspergillus fumigatus | … | … | … | … | |
HHV6 | Pneumonitis | Nivolumab (3 months) | NSCLC | Foukas et al [15] | |
SARS-CoV-2 | Pneumonitis | Nivolumab (4 months) | Metastatic renal cell carcinoma | Artigas et al [16] | |
SARS-CoV-2 | Pneumonitis | Pembrolizumab (13 cycles) | Metastatic Merkel cell carcinoma | da Costa et al [17] | |
Corynebacterium striatum | Pneumonitis | Pembrolizumab (3 cycles) | Lung adenocarcinoma | Liu et al [3] | |
P jirovecci | Pneumonitis | Pembrolizumab (11 cycles) | Refractory PMBCL | Si et al [30] | |
Gastrointestinal tract | CMV | Gastritis | Atezolizumab (8 cycles), pembrolizumab (5 cycles) | Metastatic colon and bladder cancer | Lu et al [6] |
CMV | Colitis | Pembrolizumab (9 cycles) | Metastatic melanoma | Kim et al [7] | |
CMV + Campylobacter spp | Colitis | Ipilimumab (anti-CD4) | Metastatic melanoma | Bossa et al [8] | |
Clostridioides difficile | Colitis | Durvalumab + tremelimumab (3 months) | Metastatic lung adenocarcinoma | Babacan and Tanvetyanon [27] | |
HBV | Hepatitis | Camrelizumab (3 weeks) | NPC | Zhang et al [21] | |
HBV | Hepatitis | Camrelizumab (16 weeks) | NPC | … | |
HBV | Hepatitis | Nivolumab (12 weeks) | HHC | … | |
HBV | Hepatitis | Toripalimab (35 weeks) | HNSCC | … | |
Liver | HBV | Hepatitis | Nivolumab (20 weeks) | Soft tissue carcinoma | … |
HBV | Hepatitis | Ipilimumab, nivolumab (4 cycles) | Melanoma | Koksal et al [23] | |
HBV | Hepatitis | Nivolumab (1 months) | Lung cancer | Lake [24] | |
HBV | Hepatitis | Pembrolizumab (1 cycle) | Metastatic lung adenocarcinoma | Pandey et al [25] | |
HBV | Hepatitis | Durvalumab (1 cycle) | Lung adenocarcinoma | Godbert et al [26] | |
Skin | VZV | Dermatitis | Nivolumab (6 months) | Lung adenocarcinoma | Gozzi et al [12] |
Blastomyces dermatitidis | Dermatitis (systemic infection) | Pembrolizumab (4 cycles) | Metastatic melanoma | Ferguson et al [31] | |
Malassezia spp | Dermatitis | Pembolizumab followed by ipilimumab (30 weeks) | Metastatic melanoma | Li et al [32] | |
Eye | Treponema pallidum | Anterior uveitis | Nivolumab (5 cycles) | NSCLC | Ferreira et al [29] |
Abbreviations: CMV, cytomegalovirus; CNS, central nervous system; EBV, Epstein-Barr virus; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HHV6, human herpes virus 6; HNSCC, head and neck squamous cell carcinoma; ICI, immune checkpoint inhibitor; NA, not available; NPC, nasopharyngeal carcinoma; NSCLC, nonsmall cell lung cancer; PMBCL, primary mediastinal B-cell lymphoma; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; VZV, varicella zoster virus.
Viral Infections
Several case reports document reactivation of viruses from the herpesvirus family in patients on ICIs as irAEs [6–15]. Cytomegalovirus (CMV) was shown to simulate refractory gastrointestinal autoimmune colitis or gastritis in patients without previous immunosuppressants, and only biopsy was able to differentiate these entities [6–8]. These infections fully resolved with ganciclovir. Likewise, varicella-zoster virus (VZV) reactivation involving the central nervous system (CNS), causing encephalitis, cerebral vasculopathy and atypical Ramsay-Hunt syndrome followed with ataxic sensory neuropathy [9–11], and also granulomatous dermatitis simulating ICI-autoimmune effects have also been anecdotally reported [12]. In all cases, the subsequent appearance of a typical vesicular painful rash was the clue, and diagnosis was made with the detection of VZV deoxyribonucleic acid (DNA) in the cerebrospinal fluid (CSF) by polymerase chain reaction (PCR) for the CNS manifestations, and by skin biopsy in VZV dermatitis. All patients were cured with acyclovir therapy. One case of Epstein-Barr (EBV)-induced acute cerebellar ataxia (confirmed with positive EBV DNA in the blood and CSF), and a case of fatal encephalitis, possibly in combination with concomitant pembrolizumab neurotoxicity have been reported [13, 14]. Finally, a single case of biopsy-proven human herpes virus 6 pneumonitis [15] and 2 cases of pneumonia due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strictly simulating ICI-induced pneumonitis have been reported [16–18].
Patients with hematological malignancies are at heightened risk of prolonged SARS-CoV-2 shedding and sustained pulmonary inflammation [19]. Some of the coronavirus disease 2019 (COVID-19) symptoms may simulate ICI-related pneumonitis. Distinguishing between both conditions is crucial, albeit challenging, and may require additional invasive diagnostic procedures such as bronchoscopy. Although testing and nasal screening of SARS-CoV-2 virus before anticancer therapies initiation is largely standard in many oncologic centers, there is a relatively high rate (15%) of false-negative reverse-transcription PCR results [20]. Therefore, it is possible that patients with COVID-19 are erroneously thought to have ICI-related pneumonitis.
Hepatitis B (HBV) and hepatitis C virus (HCV) infection/reactivation in patients on ICIs, although rare (<1.5%), have been reported and can simulate ICI-autoimmune hepatitis [21]. However, the risk of HBV or HBV reactivation during treatment with ICIs is still unclear, because patients with chronic hepatitis infections are excluded from randomized clinical trials with ICIs. Therefore, the natural history of preexisting HBV and HCV is variable. In some patients, the viral load regresses, suggesting that ICIs may play a role in viral clearance. In this regard, it has been observed that patients with chronic HBV infection recover the function of HBV-specific CD8+ T cells after ICI treatment [22]. Few cases of HBV reactivation have been reported in patients treated with immunotherapy and no additional immunosuppressants [21, 23–26]. Zhang et al [21] reported that among 114 patients with hepatitis B surface antigen-positive who were receiving anti-PD1/PD-L1 agents, 6 (5.3%) developed HBV reactivation, at a median of 18 weeks from ICI initiation, and 5 developed hepatitis. The risk of HBV reactivation was 17 times higher in patients without HBV prophylaxis (17.2% vs 1.2%; odds ratio = 17.50; P = .004). Four additional cases have been reported with the use of different ICIs, mainly due to lack of HBV diagnosis at baseline [23–26]. Two patients reactivated the infection after a single dose of immunotherapy, and one of them presented with fatal multiorgan failure [25, 26].
Bacterial Infections
Babacan et al [27] described 4 cases of superimposed Clostridioides difficile-associated diarrhea (CDAD) in patients with ICI-related colitis, and an additional CDAD case that developed without previous or concurrent treatment with steroids and antibiotics. In this case, CDAD preceded ICI-colitis by 2 months. Other cases of bacterial colitis simulating ICI colitis include 2 cases of Aeromonas hydrophila infection [28] and 1 case of Campylobacter spp and CMV coinfection [3].
A case of bilateral granulomatous anterior uveitis attributable to anti-PD-1 immunotherapy in a human immunodeficiency virus-positive patient with neurosyphilis has been reported [29]. The patient developed confusion with hallucinations before the fifth infusion of nivolumab. The Treponema pallidum hemagglutination assay became positive, and the lumbar puncture showed lymphocytic meningitis with no tumor cells. The clinical course was favorable with intravenous penicillin G. It was unclear whether granulomatous reaction was triggered by nivolumab in the setting of indolent syphilis.
Finally, a case of pneumonia due to Corynebacterium striatum was diagnosed in a patient receiving pembrolizumab who was unresponsive to therapy with corticosteroids due to suspected pulmonary toxicity. Bronchoscopy was crucial for diagnosis, and the patient improved with antibiotics [3].
Fungal Infections
Although exacerbation of preexisting Aspergillosis simulating cancer progression in the setting of ICI treatment has been reported (Supplementary Material References 9–11), fungi have been simulators of ICI immunotoxicity in only few instances. Specifically, Pneumocystis jirovecii pneumonia simulating ICI pneumonitis and Blastomyces dermatitidis and Malassezia spp (pityriasis versicolor) simulating ICP's dermatitis [3, 30–32].
DISCUSSION
We found that a variety of herpesviruses, and to a lesser degree hepatitis B and C viruses, SARS-CoV-2, gastrointestinal bacteria, and few fungi, can occasionally simulate irAEs, so a high index of suspicion is needed. Our review was limited by the fact that reported infections simulating ICI toxicity were case reports, subject to publication biases. Because there was no population-based assessment nor a case control design of such reports, the frequency and the spectrum of the infections simulating ICI autoimmune effects is unknown. Specifically, it is unclear whether these reported infections are merely coincidental or whether their reactivation is the bystander effect of an immunoregulatory change caused by ICI. This is an area for future prospective registries and a fertile area for future clinical research in the field of infection, immunity, and ICI treatment in modern oncology.
Furthermore, the evaluation (type and cost effectiveness) to rule out an occult infection in patients presenting with irAEs has not been validated. At present, such evaluation needs to be performed following a syndromic approach, based on atypical features of presumed irAEs and suspicion of infection. Table 2 depicts a suggested approach.
Table 2.
Organ-Specific Consideration | Work up |
---|---|
Meningoencephalitis | Assess for immunosuppression Brain MRI w/wo contrast + pituitary protocol CSF examination indicated including opening pressure CSF studies for cell count, protein, glucose, NAAT meningoencephalitis panel (including PCR for HSV and other viral PCRs), Gram stain and culture, AFB smear and culture, fungi smear and culture, cryptococcal antigen |
Pneumonitis | Assess for immunosuppression Obtain nasal respiratory viral NAAT panel (that includes SARS-CoV-2) Sputum Gram stain and culture Blood culture TB spot Serologic testing for endemic fungi Imaging studies Bronchoscopy plus bronchoalveolar lavage +/− transbronchial biopsy |
Hepatitis | Viral hepatitis (HBV, HCV, HAV, and HEV if risk factors) Liver ultrasound Consider testing for CMV, HSV, HHV6, adenoviruses, enteroviruses and Leptospirosis if clinically suspected |
Colitis | NAAT for enteropathogens including Clostridioides difficile with reflex EIA for toxin A & B Stool O&P CMV PCR from biopsy (extrapolating from IBD) Calprotectin, lactoferrin CT scan of abdomen and pelvis Consider GI endoscopy with biopsy |
Dermatitis | Assess for cellulitis Screen for HSV and Mycoplasma pneumoniae in case of erythema multiforme Skin HSV and VZV DNA in case of bullous lesions +/− Skin biopsy plus culture with specific stains (AFB, H&E, and GMS) in severe cases |
Endocrine toxicity (adrenal insufficiency) | Assess for immunosuppression Evaluate for infectious adrenalitis TB spot Serologic testing for endemic mycoses CMV testing Biopsy if clinically indicated |
Hematologic toxicity (hemolytic anemia or aplastic anemia) | Assess for immunosuppression Testing for viral and bacterial causes (Mycoplasma pneumoniae; Parvovirus B19; CMV; HHV6; EBV and HIV) Cryoglobulin analysis Screen for Shiga toxin and Escherichia coli 0157 if diarrhea and HUS Screen for HCV, HBV, HIV, and Helicobacter pylori if ITP |
Abbreviation, AFB, acid-fast bacillus; CMV, cytomegalovirus; CSF, cerebrospinal fluid; CT, computed tomography; DNA, deoxyribonucleic acid; EBV, Epstein-Barr virus; EIA, enzyme immunoassay; GI, gastrointestinal; GMS, Grocott methenamine silver; HAV, hepatitis A virus; HBV, hepatitis B virus; HCV, hepatitis C virus; H&E, hematoxylin and eosin; HEV, hepatitis E virus; HIV, human immunodeficiency virus; HHV6, human herpesvirus 6; HSV, herpes simplex virus; HUS, hemolytic uremic syndrome; IBD, inflammatory bowel disease; irAE, immune-related adverse event; ITP, immune thrombocytopenia; MRI, magnetic resonance imaging; MRSA, methicillin-resistant Staphylococcus aureus; NAAT, nucleic acid amplification test; O&P, ova and parasites; PCR, polymerase chain reaction; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TB, tuberculosis; VZV, varicella zoster virus.
NOTE: Consider infectious diseases consultation.
Some other future questions are whether these are pathogen-, host-, cell-, organ, and context-specific characteristics and whether these infections predispose to subsequent irAEs and/or influence their severity and frequency. Finally, future studies are needed to examine the questions of when to start ICIs after an infection simulating irAEs and whether specific oncological treatments predispose to develop atypical infections simulating irAEs. Because ICI use is rapidly increasing, we hope that our review will stimulate further activity in this area for future clinical research in the field of infection, immunity, and ICI treatment in modern oncology.
Supplementary Material
Acknowledgments
D. P. K. acknowledges the Robert C. Hickey Endowment and C. G. acknowledges the Instituto de Salud Carlos III, Subdirección General de Redes y Centros de Investigación Cooperativa, Ministerio de Economía, Industria y Competitividad, Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC).
Potential conflicts of interest. D. P. K. reports honoraria and research support from Gilead Sciences and Astellas, Inc., received consultant fees from Astellas Pharma, Merck, and Gilead Sciences, and is a member of the Data Review Committee of Cidara Therapeutics, AbbVie, and the Mycoses Study Group. C. G. reports honoraria and research support from Pfizer and Merck international and consulting fees from Gilead. P. C. O. reports grant or research support from Merck Sharp & Dohme Corp., Deinove Pharmaceuticals, Summit Pharmaceuticals, and Melinta Pharmaceuticals and consulting fees from Ferring Pharmaceuticals Inc. and Napo Pharmaceuticals. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.
Contributor Information
Carlota Gudiol, Infectious Diseases Department, Bellvitge University Hospital, IDIBIELL, University of Barcelona, Barcelona, Spain; Institut Català d’Oncologia, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), (CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain.
Rachel S Hicklen, Research Medical Library, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Pablo C Okhyusen, Division of Infectious Diseases, LSU Health Shreveport, Shreveport, Louisiana, USA.
Alexandre E Malek, Division of Infectious Diseases, LSU Health Shreveport, Shreveport, Louisiana, USA.
Dimitrios P Kontoyiannis, Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Supplementary Data
Supplementary materials are available at Open Forum Infectious Diseases online. Consisting of data provided by the authors to benefit the reader, the posted materials are not copyedited and are the sole responsibility of the authors, so questions or comments should be addressed to the corresponding author.
References
- 1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12:252–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Xu C, Chen Y-P, Du X-J, et al. Comparative safety of immune checkpoint inhibitors in cancer: systematic review and network meta-analysis. BMJ 2018; 7:k4226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Liu Z, Liu T, Zhang X, et al. Opportunistic infections complicating immunotherapy for non-small cell lung cancer. Thorac Cancer 2020; 11:1689–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Morelli T, Fujita K, Redelman-Sidi G, Elkington PT. Infections due to dysregulated immunity: an emerging complication of cancer immunotherapy. Thorax 2022; 77:304–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Barber DL, Mayer-Barber KD, Feng CG, et al. CD4 T cells promote rather than control tuberculosis in the absence of PD-1—mediated inhibition. J Immunol 2011; 186:1598–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Lu J, Firpi-Morell RJ, Dang LH, et al. An unusual case of gastritis in one patient receiving PD-1 blocking therapy: coexisting immune-related gastritis and cytomegaloviral infection. Gastroenterology Res 2018; 11:383–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Kim H, Ha SY, Kim J, Kang M, Lee J. Severe cytomegalovirus gastritis after pembrolizumab in a patient with melanoma. Curr Oncol 2020; 27:e436–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bossa F, Perri F, Niro G, Parente P, Graziano P, Andriulli A. Cytomegalovirus colitis in a patient treated with ipilimumab for metastatic melanoma. Aliment Pharmacol Ther 2016; 43:174–5. [DOI] [PubMed] [Google Scholar]
- 9. Watanabe Y, Kikuchi R, Iwai Y, et al. Varicella zoster virus encephalitis mimicking nivolumab-induced autoimmune neuropathy in a patient with lung cancer. J Thorac Oncol 2019; 14:e163–5. [DOI] [PubMed] [Google Scholar]
- 10. Ursu R, Roumi A, Chouahnia K, Altmayer V, Cuzzubbo S, Carpentier AF. Varicella zoster virus vasculopathy in a patient treated with immune checkpoint inhibitor for lung cancer. Rev Neurol (Paris) 2019; 175:95–7. [DOI] [PubMed] [Google Scholar]
- 11. Sakoh T, Kanzaki M, Miyamoto A, et al. Ramsay-Hunt syndrome and subsequent sensory neuropathy as potential immune-related adverse events of nivolumab: a case report. BMC Cancer 2019; 19:1220–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Gozzi E, Rossi L, Angelini F, et al. Herpes zoster granulomatous dermatitis in metastatic lung cancer treated with nivolumab: a case report. Thorac Cancer 2020; 11:1330–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Saikawa H, Nagashima H, Maeda T, Maemondo M. Acute cerebellar ataxia due to Epstein–Barr virus under administration of an immune checkpoint inhibitor. BMJ Case Rep 2019; 12:e231520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Johnson DB, McDonnell WJ, Gonzalez-Ericsson PI, et al. A case report of clonal EBV-like memory CD4+ T cell activation in fatal checkpoint inhibitor-induced encephalitis. Nat Med 2019; 25:1243–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Foukas PG, Tsiodras S, Economopoulou P, et al. Concomitant human herpes virus 6 and nivolumab-related pneumonitis: potential pathogenetic insights. ID Cases 2018; 11:101–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Artigas C, Lemort M, Mestrez F, Gil T, Flamen P. COVID-19 pneumonia mimicking immunotherapy-induced pneumonitis on 18f-FDG PET/CT in a patient under treatment with nivolumab. Clin Nucl Med 2020; 45:e381–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. da Costa CM, de Souza ZS, Real Salgues AC, et al. COVID-19 in a patient with advanced Merkel cell carcinoma receiving immunotherapy. Immunotherapy 2020; 12:1133–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Dipasquale A, Persico P, Lorenzi E, Rahal D, Santoro A, Simonelli M. COVID-19 lung injury as a primer for immune checkpoint inhibitors (ICIs)-related pneumonia in a patient affected by squamous head and neck carcinoma treated with PD-L1 blockade: a case report. J Immunother Cancer 2021; 9:e001870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Abdul-Jawad S, Baù L, Alaguthurai T, et al. Acute immune signatures and their legacies in severe acute respiratory syndrome coronavirus-2 infected cancer patients. Cancer Cell 2021; 39:257–275.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Niu A, Ning B, Socola F, et al. High mortality with high false negative rate: COVID-19 infection in patients with hematologic malignancies. Leuk Res 2021; 106:106582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Zhang X, Zhou Y, Chen C, et al. Hepatitis B virus reactivation in cancer patients with positive hepatitis B surface antigen undergoing PD-1 inhibition. J Immunother Cancer 2019; 7:322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. De Keukeleire SJ, Vermassen T, Nezhad ZM, et al. Managing viral hepatitis in cancer patients under immune checkpoint inhibitors: should we take the risk? Immunotherapy 2021; 13:409–18. [DOI] [PubMed] [Google Scholar]
- 23. Koksal AS, Toka B, Eminler AT, et al. HBV-related acute hepatitis due to immune checkpoint inhibitors in a patient with malignant melanoma. Ann Oncol 2017; 28:3103–4. [DOI] [PubMed] [Google Scholar]
- 24. Lake AC. Hepatitis B reactivation in a long-term nonprogressor due to nivolumab therapy. AIDS 2017; 31:2115–8. [DOI] [PubMed] [Google Scholar]
- 25. Pandey A, Ezemenari S, Liaukovich M, et al. A rare case of pembrolizumab-induced reactivation of hepatitis B. Case Rep Oncol Med 2018; 2018:1–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Godbert B, Petitpain N, Lopez A, Nisse YE, Gillet P. Hepatitis B reactivation and immune check point inhibitors. Dig Liver Dis 2021; 53:452–5. [DOI] [PubMed] [Google Scholar]
- 27. Babacan NA, Tanvetyanon T. Superimposed clostridium difficile infection during checkpoint inhibitor immunotherapy-induced colitis. J Immunother 2019; 42:350–3. [DOI] [PubMed] [Google Scholar]
- 28. Yamauchi Y, Arai M, Akizue N, et al. Colonoscopic evaluation of diarrhea/colitis occurring as an immune-related adverse event. Jpn J Clin Oncol 2021; 51:363–70. [DOI] [PubMed] [Google Scholar]
- 29. Ferreira M, Bastides F, Pichon E, Lisee F, Marchand-Adam S, Flament T. Neurological abnormalities and syphilitic serologic variation with nivolumab: a case of neurosyphilis? Eur Respiratory J Conf 2020; 56:1745. [Google Scholar]
- 30. Si S, Erickson K, Evageliou N, Silverman M, Kersun L. A usual presentation of pneumocystis jirovecii pneumonia in a woman treated with immune checkpoint inhibitor. J Pediatr Hematol Oncol 2021; 43:e163–4. [DOI] [PubMed] [Google Scholar]
- 31. Ferguson I, Heberton M, Compton L, Keller J, Cornelius L. Disseminated blastomycosis in a patient on pembrolizumab for metastatic melanoma. JAAD Case Rep 2019; 5:580–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Li M, Spaccarelli N, Kendra K, Wu RC, Verschraegen C. Refractory dermatitis contributed by pityriasis versicolor: a case report. J Med Case Rep 2021; 15:212. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.