Next Article in Journal
Biological Control of a Root-Knot Nematode Meloidogyne incognita Infection of Tomato (Solanum lycopersicum L.) by the Oomycete Biocontrol Agent Pythium oligandrum
Previous Article in Journal
Habitat Diversity, Environmental Conditions, and Distribution of Endangered Fungus Sarcosoma globosum (Ascomycota) in Lithuania
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Disseminated Histoplasmosis in a Patient with Myelofibrosis on Ruxolitinib: A Case Report and Review of the Literature on Ruxolitinib-Associated Invasive Fungal Infections

by
Chia-Yu Chiu
1,
Teny M. John
1,
Takahiro Matsuo
1,
Sebastian Wurster
1,
Rachel S. Hicklen
2,
Raihaan Riaz Khattak
1,
Ella J. Ariza-Heredia
1,
Prithviraj Bose
3 and
Dimitrios P. Kontoyiannis
1,*
1
Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
2
Research Medical Library, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
3
Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
J. Fungi 2024, 10(4), 264; https://doi.org/10.3390/jof10040264
Submission received: 24 February 2024 / Revised: 24 March 2024 / Accepted: 29 March 2024 / Published: 31 March 2024

Abstract

:
Ruxolitinib, a selective inhibitor of Janus kinases, is a standard treatment for intermediate/high-risk myelofibrosis (MF) but is associated with a predisposition to opportunistic infections, especially herpes zoster. However, the incidence and characteristics of invasive fungal infections (IFIs) in these patients remain uncertain. In this report, we present the case of a 59-year-old woman with MF who developed disseminated histoplasmosis after seven months of ruxolitinib use. The patient clinically improved after ten weeks of combined amphotericin B and azole therapy, and ruxolitinib was discontinued. Later, the patient received fedratinib, a relatively JAK2-selective inhibitor, without relapse of histoplasmosis. We also reviewed the literature on published cases of proven IFIs in patients with MF who received ruxolitinib. Including ours, we identified 28 such cases, most commonly due to Cryptococcus species (46%). IFIs were most commonly disseminated (39%), followed by localized lung (21%) infections. Although uncommon, a high index of suspicion for opportunistic IFIs is needed in patients receiving JAK inhibitors. Furthermore, the paucity of data regarding the optimal management of IFIs in patients treated with JAK inhibitors underscore the need for well-designed studies to evaluate the epidemiology, pathobiology, early diagnosis, and multimodal therapy of IFIs in patients with hematological malignancies receiving targeted therapies.

1. Introduction

Myelofibrosis (MF) is the most advanced chronic Philadelphia chromosome-negative myeloproliferative neoplasm (MPN). It may present as primary myelofibrosis (PMF) or evolve from essential thrombocythemia (ET; post-ET MF) or polycythemia vera (PV; post-PV MF) [1]. Ruxolitinib, an orally bioavailable potent and selective inhibitor of Janus kinases (JAK) 1 and 2, has been approved for the treatment of MF and PV [2]. Ruxolitinib has been shown to reduce spleen size, decrease symptoms, and improve survival in patients with MF [3].
However, ruxolitinib exerts potent anti-inflammatory and immunosuppressive effects and has been associated with infectious complications, most commonly urinary tract infection (4.7–24.6%), pneumonia (5.3–13.1%), herpes zoster (1.9–11.5%), and sepsis (1.3–7.9%) [4]. Other opportunistic infections reported in these patients include tuberculosis, toxoplasmosis, progressive multifocal leukoencephalopathy, and hepatitis B reactivation [5,6]. Although opportunistic fungal infections (e.g., cryptococcosis and Pneumocystis jirovecii pneumonia) have been reported in MF patients receiving ruxolitinib [5], the incidence and characteristics of invasive fungal infections (IFIs) in these patients remain uncertain.
Herein, we present a patient with MF who developed disseminated histoplasmosis after seven months of ruxolitinib treatment. Additionally, we critically review the literature on all published cases of proven IFIs in MF patients who received ruxolitinib.

2. Case Summary

A 59-year-old woman presented to our hospital in April 2023 with one month of fever. She is originally from Pakistan and immigrated to the USA 30 years ago. She had a history of PV, was diagnosed in March 2019, and was treated with acetylsalicylic acid and hydroxyurea. In August 2022, she was diagnosed with MF on bone marrow biopsy after presenting with progressive weight loss, fatigue, early satiety, and splenomegaly (19 cm in craniocaudal dimension). Ruxolitinib was started at 10 mg twice daily and was increased to 15 mg twice daily one month later. In January 2023, the patient experienced an episode of Legionella pneumonia that was treated with levofloxacin on an outpatient basis. At the beginning of February 2023, she went to Puerto Vallarta, Mexico, for a 3-day trip and reported seeing a few bats at the hotel restaurant. At the end of February 2023, she developed persistent high-grade fever (104 °F) associated with fatigue, dry cough, weight loss, and poor appetite. She was admitted to an outside hospital and received empiric antibiotic therapy with cefepime plus doxycycline. An extensive infectious disease workup was negative, including human immunodeficiency virus, syphilis, hepatitis A, B, and C, Bartonella serology, Brucella serology, Q fever serology, typhus serology, galactomannan, (1,3)-beta-D-glucan, Cryptococcus serum antigen, cytomegalovirus plasma quantitative polymerase chain reaction (PCR), Epstein–Barr virus plasma quantitative PCR, and blood cultures. Bone marrow biopsy revealed MF with negative Gram, Giemsa, and acid-fast stains. The patient’s fever slightly improved during her hospital stay, and she was discharged with prednisone (20 mg twice daily) for anti-inflammatory purposes as her infectious disease workup was negative.
A few days later, the patient was readmitted to our hospital (MD Anderson Cancer Center, Houston, TX, USA) with severe epigastric pain and recurrent non-neutropenic fever despite prednisone. Computed tomography (CT) showed cervical lymphadenopathy, the largest lymph node being in the left subclavicular region (Figure 1A), and hepatosplenomegaly with splenic infarction (Figure 1B). At that time, an outside hospital report of a positive Histoplasma urine antigen (>20 ng/mL; MiraVista diagnostics, Indianapolis, IN, USA) and recovery of Histoplasma capsulatum from bone marrow culture was received. Repeat infectious disease workup that included Aspergillus antigen and Cryptococcus serum antigen was negative. The (1,3)-beta-D-glucan level was borderline positive at 80 pg/mL. Her urine Histoplasma antigen (Mayo Clinic Laboratories, Rochester, MN, USA) was negative. A repeat bone marrow biopsy, fungal blood culture, and cervical lymph node biopsy were performed, and all grew Histoplasma capsulatum (Figure 1C). This was identified as a member of Latin American group A by whole-genome sequencing at the Fungus Testing Laboratory, University of Texas Health Science Center in San Antonio.
Once the patient was diagnosed with disseminated histoplasmosis, intravenous (IV) liposomal amphotericin B (3 mg/kg/day) was started immediately, and itraconazole (200 mg oral capsule, twice daily) was added 5 days later. After initial improvement of the fever, the patient was discharged on this antifungal regimen. She was readmitted shortly thereafter with a recurrent fever of 102 °F and persistent abdominal pain. Positron emission tomography (PET) revealed newly developed mediastinal lymphadenopathy (Figure 2A) and stable hepatosplenomegaly. Liposomal amphotericin B was increased to 5 mg/kg/day, and oral itraconazole was switched to oral posaconazole (300 mg tablet daily). Her fever subsequently subsided, and she was discharged.
The patient subsequently developed generalized edema, acute kidney injury, and new onset hypertension while on liposomal amphotericin B and posaconazole. Therefore, the IV liposomal amphotericin B dose was decreased to 5 mg/kg three times per week. Her serum posaconazole and potassium levels were 1200 ng/mL and 3.7 mEq/L, respectively. Liposomal amphotericin B was discontinued and posaconazole was switched to isavuconazole due to concern of posaconazole-induced pseudoaldosteronism. Hypertension, weight gain, and hypokalemia abated after the switch from posaconazole to isavuconazole. Ruxolitinib was discontinued following the diagnosis of histoplasmosis in June 2023. In September 2023, the patient was started on fedratinib, a relatively JAK2-selective inhibitor approved for the treatment of MF, which was tolerated without any significant adverse events or relapse of histoplasmosis to date.
On her 10th month of antifungal therapy, the patient maintained a complete clinical and radiologic response (i.e., serial follow-up PETs showed decreased F-18 FDG avidity of lymph nodes) (Figure 2B). Multiple serum Histoplasma serologies (complement fixation and immunodiffusion assay) and urine Histoplasma antigen tests (Mayo Clinic Laboratories, MN, USA) have been negative since April 2023 (Figure 3). As of February 2024, the patient continues to receive antifungal monotherapy with isavuconazole (Figure 3).

3. Case Discussion and Systematic Review of the Literature

A comprehensive literature review was conducted to identify patients with MF who developed IFIs after receiving ruxolitinib. From inception to February 2024, OVID-Embase, OVID-MEDLINE, PubMed, and Scopus were queried using both natural language and controlled vocabulary terms for myelofibrosis, ruxolitinib, and fungal infections. The full search strategy is available in Supplementary Materials. We also searched the reference lists of all relevant publications for additional references. We included cases that provided adequate information regarding the diagnosis, treatment, and outcome of IFIs.
The reported incidence of IFIs in patients who received ruxolitinib for MF was 0.6–2.6% but increased to 3.7–4.9% in mixed cohorts of patients with ET, PV, MF, and stem cell transplant recipients who receive ruxolitinib as a treatment for graft-vs-host disease (GVHD) (Table 1).
The literature review identified 28 MF patients (including ours) who had proven IFIs while receiving ruxolitinib [5,10,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32]. The patients’ median age was 70 years (interquartile range [IQR], 65–73). Most patients were male (69%) and had primary MF (54%). The median time from ruxolitinib initiation to IFI onset was 11 months (IQR 6–27). IFIs were most commonly disseminated (39%), followed by localized lung (21%) and central nervous system (18%) infections. The most common pathogens were Cryptococcus spp. (46%), Coccidioides spp. (11%), and Pneumocystis jirovecii (11%). In most cases, ruxolitinib was discontinued permanently (76%) or put on hold (18%). No patients had received antifungal prophylaxis at IFI diagnosis. Five patients (18%) had co-occurring infections, including one case each of Klebsiella pneumoniae bacteremia, pulmonary tuberculosis, pneumonia due to Mycobacterium avium complex, and Mycobacterium haemophilum skin infection. One patient had disseminated tuberculosis and shingles (herpes zoster) (Table 2).
The mechanism of action of ruxolitinib may explain the occurrence of IFIs. Ruxolitinib inhibits JAK-1 and JAK-2 acts on downstream Signal Transducer and Activators of the Transcription (STAT) pathways. JAK-STAT pathways are crucial for the activation and differentiation of various innate (e.g., dendritic cells, natural killer cells) and adaptive (e.g., type-1 and type-17 T-helper cells) immune cell subsets that are pivotal effectors and orchestrators of antifungal host defense [4,33]. Specifically, animal studies have shown that the interferon-gamma–JAK1/2–STAT1 axis plays an essential role in the protective response against cryptococcosis via STAT1-mediated activation of classical (M1) macrophages and effective type-1 T-helper cell responses [34,35]. This might explain why Cryptococcus infection is the dominant IFI in patients who receive ruxolitinib. Furthermore, JAK1/2 inhibitors lead to decreased production of cytokines, including interleukins (e.g., IL-6), interferon-gamma, tumor necrosis factor-alpha, and granulocyte-macrophage colony-stimulating factor, and also affect downstream receptor signaling in response to cytokine stimuli (e.g., IL-2) [34,35]. These cytokines are well-known “master regulators” of antifungal defense against all classes of pathogenic fungi and have pleiotropic roles in immune cell recruitment, differentiation, and maturation, as well as the initiation of antifungal effector responses by neutrophils and mononuclear phagocytes [33]. Despite these relatively well-understood molecular mechanisms, determinants of individual IFI risk and the relevance of other factors (such as age, comorbidities, pre-existing cytopenia, previous treatments, concurrent immunosuppressive therapy, and intensity of exposure to fungi) and possible immunogenetic defects remain unclear.
The overall infection rate was not significantly different between ruxolitinib and the control groups (placebo or best available therapy) in phase 3 clinical trials of patients with various MPNs, except for a significantly higher risk of herpes zoster infection in patients receiving ruxolitinib [5]. This observation was made in both MF patients (COMFORT I and COMFORT II trials) [9,36] and PV patients (RESPONSE and RESPONSE-2 trials) [37,38]. In contrast, the largest national registry, which included 837 patients with MF in Sweden, showed an increased risk of infection with ruxolitinib compared to other cytoreductive therapies [39]. It remains controversial whether the observed increased risk of infection in MF is more inherent to the underlying hematologic disorder or related to ruxolitinib. Interestingly, we found only two published cases of PV (not included in Table 2) that developed IFIs when receiving ruxolitinib [40,41] but 28 MF patients who developed proven IFIs when receiving ruxolitinib (Table 2). However, given the limited number of IFIs reported in patients who received ruxolitinib, it remains unclear whether the underlying hematologic disease (PV vs. MF) influences the risk of IFIs.
One retrospective study showed that 123 out of 446 MF patients experienced infectious events after a median ruxolitinib exposure of 24 months [8]. In patients with MF on ruxolitinib, both a prior history of infectious events and a high international prognostic scoring system (IPSS) score were significantly correlated with higher infectious risk [8]. Interestingly, the infection rate decreased over time with ruxolitinib exposure, and the total cumulative dose of ruxolitinib was not correlated with infection risk [8]. Furthermore, spleen reduction ≥ 50% from baseline after 3 months of ruxolitinib treatment was associated with better infection-free survival [8]. These findings reinforce the concept that MF disease severity is an important risk factor for infections, and the therapeutic effect of ruxolitinib in reducing splenomegaly might reduce infection risk. Notably, 39% of the cases in our review had documented splenomegaly at the onset of IFIs, which might imply that poor oncologic response to ruxolitinib could be a critical modulator of IFI risk in these patients.
It is important to recognize that IFIs can occur at any time after ruxolitinib treatment is initiated, as indicated by the broad IQR of 6–27 months from ruxolitinib initiation to IFI onset in our review (Table 2). Thus, there has been debate about whether the degree of immunosuppression from ruxolitinib mandates any infectious disease screening or the initiation of antimicrobial prophylaxis [40,42,43]. Given the relative infrequency of recognized IFIs, we believe a more proactive approach that includes routine screening and antifungal prophylaxis should be reserved only for those with high-risk exposure or prior history of opportunistic infections, including fungal infections.
Once an IFI is diagnosed, the appropriate course of ruxolitinib therapy remains unclear. Although a few patients resumed or continued ruxolitinib [12,25,28,30], ruxolitinib was discontinued permanently in most cases (76%, Table 2). This approach was also common in patients who had tuberculosis, herpes zoster, or hepatitis B reactivation while on ruxolitinib [15,17]. Currently, there is no large-scale study about switching ruxolitinib to pacritinib (JAK 2-selective inhibitor with virtually no JAK 1 activity) or fedratinib (relatively selective for JAK 2 over JAK 1) in MF patients with IFIs. To our knowledge, our patient is the first who developed disseminated mycosis (histoplasmosis) as a complication of ruxolitinib and has, to date, not experienced a relapse of infection after switching to the more selective JAK2 inhibitor fedratinib. Although clinical data are lacking, the rationale for this switch was based on the theoretical consideration that the immunosuppressive effects of ruxolitinib stem primarily from its inhibition of JAK1 rather than JAK2. While pacritinib is even more selective for JAK2 than fedratinib, with virtually no anti-JAK1 activity, fedratinib was selected in our patient with a preserved platelet count (>100 × 109/L) because of more robust efficacy data for fedratinib in non-cytopenic MF patients [44].
Additionally, other strategies, such as adjuvant therapy with granulocyte macrophage colony-stimulating factor, interferon-gamma, or other immunomodulators [45,46], might be helpful in MF patients who need to continue JAK inhibitor therapy. There is a need for thorough studies of the impact of various immunotherapies that boost antifungal immunity in patients with hematological malignancies who have developed IFIs while receiving targeted therapies [47].
In recent years, the epidemiology of IFI in cancer patients has changed significantly, influenced by advances in antineoplastic therapies, improved early detection and identification methods (such as positron emission tomography, matrix-assisted laser desorption ionization-time of flight mass spectroscopy, and whole-genome sequencing), and the introduction of novel antifungal agents for both treatment and prophylaxis [48,49]. New immunotherapeutic and molecularly targeted agents acting on specific molecular pathways are generally better tolerated and more effective than traditional chemotherapy [11]. However, the downstream consequences of inhibiting these pathways may not be fully understood in randomized controlled trials in view of the patient selection and the fact such trials exclude patients with active infections. As a result, significant or unique infectious complications, including IFIs, may only become apparent when these therapies are administered to larger, more diverse populations beyond the confines of clinical trials. This poses substantial challenges in assessing the individual risk associated with each therapeutic agent. Therefore, it is imperative to deepen our understanding of each agent’s specific risks for developing IFIs and the natural history of these infections in patients with hematologic malignancies receiving various targeted therapies. A more standardized approach for capturing IFIs with immunogenetic and immunophenotyping strategies would allow for the accurate assessment of infection risk in individual patients and provide personalized approaches to prevent opportunistic fungal diseases associated with molecularly targeted agents [50].
In the United States, Histoplasma capsulatum was previously considered highly endemic to the Ohio and Mississippi River Valleys, a notion established in the 1970s based on antigen skin testing [51]. However, studies in the 2020s have shown that the distribution of histoplasmosis extends beyond these historical boundaries [51]. The expansion of geographic distribution over the past half-century can be attributed to multiple factors, including improvements in diagnostic methods, increased travel, climate change, and changes in land development practices [51,52,53]. Although travel-related fungal infections have often been detected in outbreak settings rather than through routine surveillance, travel-related exposure is considered a significant contributor to the occurrence of dimorphic mycoses [51,54]. In our case, travel to Puerto Vallarta, Mexico, a renowned bat-watching site, is the most likely source of the patient’s Histoplasma capsulatum exposure rather than her residence in Texas. Our patient’s isolate belonged to the Latin American group A (LAm A), the predominant clade in Latin America [52]. This contrasts with the United States, where the common clades of Histoplasma capsulatum are North American Classes 1 and 2 (NAm1 and NAm2) [52]. The use of whole-genome sequencing for fungal identification to the clade level will help clinicians better understand species distribution and confirm travel-related fungal infections [55]. Histoplasma capsulatum grew in soil enriched with bat guano and may even infect bats [56]. Exposures occurred during activities such as construction, renovation, demolition, soil excavation, spelunking, and cleaning sites harboring the fungus [54,57].
There are several important teaching points in our patient’s diagnosis and management of histoplasmosis. First, the sensitivity of the Histoplasma urine antigen tests varies across different assays, and the assay used in our hospital yielded a false-negative result [58]. In addition, our patient also did not elicit Histoplasma serum antibodies, which could be attributed to impaired humoral and T-cell immunity resulting from ruxolitinib [59]. Therefore, the borderline positive 1,3-beta-d-glucan was the only positive serum fungal biomarker for an IFI in our case, consistent with reports that Histoplasma, as many fungi, can produce 1,3-beta-d-glucan [60]. Second, our patient possibly developed posaconazole-induced pseudohyperaldosteronism [61,62]. However, we did not perform laboratory testing (11-deoxycortisol, plasma aldosterone, and renin activity) for confirmation. Itraconazole and posaconazole are more commonly associated with pseudohyperaldosteronism than fluconazole, voriconazole, and isavuconazole [63]. In our institution, we previously reported a patient who developed posaconazole-induced pseudohyperaldosteronism, in whom the symptoms resolved upon switching to isavuconazole [62]. Third, itraconazole is the recommended step-down therapy for disseminated histoplasmosis following initial improvements with amphotericin B [64]. However, our patient developed worsening symptoms and new mediastinal lymphadenopathy during treatment with itraconazole. Subsequently, the patient exhibited clinical improvement with isavuconazole, administered concurrently with fedratinib. Although isavuconazole appears to be a safe and effective treatment for histoplasmosis [65], the data supporting its superiority over itraconazole in patients receiving JAK inhibitors are lacking. Antifungal susceptibility of Histoplasma capsulatum was not tested in this case. Although in vitro antifungal susceptibility testing for dimorphic fungi remains unstandardized. Although epidemiological cut-off values have been proposed, no clinical susceptibility breakpoints have been determined for these organisms [66]. Few in vitro studies have demonstrated low minimum inhibitory concentrations for most mold-active triazoles, including isavuconazole against Histoplasma capsulatum [67,68,69]. Fourth, we do not have in-hospital guidelines for treating disseminated histoplasmosis in cancer patients, but we follow the Infectious Diseases Society of America clinical practice guidelines for the management of patients with histoplasmosis with some modifications in cancer patients [64]. We usually start cancer patients with disseminated and serious fungal infections on combination therapy with liposomal amphotericin B and an azole until therapeutic azole levels are reached.
Our review has several limitations. First, only proven IFI cases were analyzed in-depth in this study, which underestimates the incidence of IFIs in patients who received ruxolitinib. However, at the time of this review, no study focused on patients with possible or probable IFIs who received ruxolitinib. Second, ruxolitinib is used to treat MF/PV and GVHD. In this review, we did not discuss stem cell transplant recipients who received ruxolitinib as part of GVHD management because these patients were usually previously exposed to or concomitantly received other immunosuppressants. For instance, we found one case of disseminated aspergillosis in a patient with acute myeloid leukemia who underwent hematopoietic stem cell transplantation and received tacrolimus, ruxolitinib, and budesonide for GVHD [70]. Third, whether ruxolitinib-associated IFIs are time-dependent, dose-dependent, or neither remains to be seen. Well-designed studies or registries are warranted to evaluate the epidemiology and risk of ruxolitinib-associated IFIs. Fourth, IPSS or other MF prognostic scores were unavailable in the reviewed cases, limiting our ability to examine the correlation between IFIs and MF severity.
In summary, although uncommon, IFIs can complicate ruxolitinib therapy for MF, a hematologic disease historically considered to be associated with very low risk of IFIs in the pre-JAK inhibitor era [39]. Thus, physicians should have a high index of suspicion for potential IFIs in patients on ruxolitinib to facilitate early diagnosis and initiation of appropriate antifungal therapy. Screening and prophylaxis for fungal infections in patients receiving ruxolitinib should be considered based on patient-specific factors. Furthermore, the knowledge gaps identified in our review underscore the critical global need to study the epidemiology, pathobiology, early diagnosis, and multimodal therapy of IFIs in patients with hematological malignancies receiving targeted therapies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jof10040264/s1.

Author Contributions

Conceptualization, D.P.K.; methodology, C.-Y.C. and R.S.H.; software, C.-Y.C.; validation, C.-Y.C., T.M.J., T.M., S.W. and R.S.H.; formal analysis, C.-Y.C.; data curation, C.-Y.C. and R.S.H.; writing—original draft preparation, C.-Y.C. and S.W.; writing—review and editing, T.M.J., T.M., R.R.K., E.J.A.-H., P.B. and D.P.K.; visualization, C.-Y.C., T.M. and S.W.; supervision, D.P.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Case details have been anonymized, and the patient signed a consent form permitting publication of this case and clinical images.

Data Availability Statement

The de-identified datasets generated during and/or analyzed during the current study are available from the corresponding author upon reasonable request. The data are not publicly available due to ethical reasons.

Conflicts of Interest

D.P.K. reports honoraria and research support from Gilead Sciences and Astellas Pharma. He received consultant fees from Astellas Pharma, Merck, and Gilead Sciences and is a member of the Data Review Committee of Cidara Therapeutics, AbbVie, Scynexis, and the Mycoses Study Group. T.M.J receives research support from Illumina Inc. All other authors declare no potential conflicts of interest.

References

  1. Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef]
  2. JAKAFI (Ruxolitinib) Label. U.S. Food and Drug Administration. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/202192s023lbl.pdf (accessed on 10 February 2024).
  3. Verstovsek, S.; Gotlib, J.; Mesa, R.A.; Vannucchi, A.M.; Kiladjian, J.J.; Cervantes, F.; Harrison, C.N.; Paquette, R.; Sun, W.; Naim, A.; et al. Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J. Hematol. Oncol. 2017, 10, 156. [Google Scholar] [CrossRef] [PubMed]
  4. Elli, E.M.; Baratè, C.; Mendicino, F.; Palandri, F.; Palumbo, G.A. Mechanisms Underlying the Anti-inflammatory and Immunosuppressive Activity of Ruxolitinib. Front. Oncol. 2019, 9, 1186. [Google Scholar] [CrossRef]
  5. Lussana, F.; Cattaneo, M.; Rambaldi, A.; Squizzato, A. Ruxolitinib-associated infections: A systematic review and meta-analysis. Am. J. Hematol. 2018, 93, 339–347. [Google Scholar] [CrossRef] [PubMed]
  6. Peng, Y.; Meng, L.; Hu, X.; Han, Z.; Hong, Z. Tuberculosis in Patients with Primary Myelofibrosis During Ruxolitinib Therapy: Case Series and Literature Review. Infect. Drug Resist. 2020, 13, 3309–3316. [Google Scholar] [CrossRef] [PubMed]
  7. Polverelli, N.; Breccia, M.; Benevolo, G.; Martino, B.; Tieghi, A.; Latagliata, R.; Sabattini, E.; Riminucci, M.; Godio, L.; Catani, L.; et al. Risk factors for infections in myelofibrosis: Role of disease status and treatment. A multicenter study of 507 patients. Am. J. Hematol. 2017, 92, 37–41. [Google Scholar] [CrossRef]
  8. Polverelli, N.; Palumbo, G.A.; Binotto, G.; Abruzzese, E.; Benevolo, G.; Bergamaschi, M.; Tieghi, A.; Bonifacio, M.; Breccia, M.; Catani, L.; et al. Epidemiology, outcome, and risk factors for infectious complications in myelofibrosis patients receiving ruxolitinib: A multicenter study on 446 patients. Hematol. Oncol. 2018, 36, 561–569. [Google Scholar] [CrossRef]
  9. Verstovsek, S.; Mesa, R.A.; Gotlib, J.; Gupta, V.; DiPersio, J.F.; Catalano, J.V.; Deininger, M.W.; Miller, C.B.; Silver, R.T.; Talpaz, M.; et al. Long-term treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. J. Hematol. Oncol. 2017, 10, 55. [Google Scholar] [CrossRef]
  10. Kusne, Y.; Kimes, K.E.; Feller, F.F.; Patron, R.; Banacloche, J.G.; Blair, J.E.; Vikram, H.R.; Ampel, N.M. Coccidioidomycosis in Patients Treated with Ruxolitinib. Open Forum Infect. Dis. 2020, 7, ofaa167. [Google Scholar] [CrossRef]
  11. Gold, J.A.W.; Tolu, S.S.; Chiller, T.; Benedict, K.; Jackson, B.R. Incidence of Invasive Fungal Infections in Patients Initiating Ibrutinib and Other Small Molecule Kinase Inhibitors-United States, July 2016–June 2019. Clin. Infect. Dis. 2022, 75, 334–337. [Google Scholar] [CrossRef]
  12. Wysham, N.G.; Sullivan, D.R.; Allada, G. An opportunistic infection associated with ruxolitinib, a novel janus kinase 1,2 inhibitor. Chest 2013, 143, 1478–1479. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, C.C.; Chen, Y.Y.; Huang, C.E. Cryptococcal meningoencephalitis associated with the long-term use of ruxolitinib. Ann. Hematol. 2016, 95, 361–362. [Google Scholar] [CrossRef] [PubMed]
  14. Hirano, A.; Yamasaki, M.; Saito, N.; Iwato, K.; Daido, W.; Funaishi, K.; Ishiyama, S.; Deguchi, N.; Taniwaki, M.; Ohashi, N. Pulmonary cryptococcosis in a ruxolitinib-treated patient with primary myelofibrosis. Respir. Med. Case Rep. 2017, 22, 87–90. [Google Scholar] [CrossRef] [PubMed]
  15. Dioverti, M.V.; Abu Saleh, O.M.; Tande, A.J. Infectious complications in patients on treatment with Ruxolitinib: Case report and review of the literature. Infect. Dis. 2018, 50, 381–387. [Google Scholar] [CrossRef] [PubMed]
  16. Tsukui, D.; Fujita, H.; Suzuki, K.; Hirata, K. A case report of cryptococcal meningitis associated with ruxolitinib. Medicine 2020, 99, e19587. [Google Scholar] [CrossRef]
  17. Ogai, A.; Yagi, K.; Ito, F.; Domoto, H.; Shiomi, T.; Chin, K. Fatal Disseminated Tuberculosis and Concurrent Disseminated Cryptococcosis in a Ruxolitinib-treated Patient with Primary Myelofibrosis: A Case Report and Literature Review. Intern. Med. 2022, 61, 1271–1278. [Google Scholar] [CrossRef] [PubMed]
  18. Harvey, J.; Tran, L.; Sampath, R.; White, C.; Campanile, T. 1410. Serious Cryptococcal Infections with Ruxolitinib Use: A Case of Meningitis and a Review of the Literature. Open Forum Infect. Dis. 2019, 6 (Suppl. S2), S513–S514. [Google Scholar]
  19. Ciochetto, Z.; Wainaina, N.; Graham, M.B.; Corey, A.; Abid, M.B. Cryptococcal infection with ruxolitinib in primary myelofibrosis: A case report and literature review. Clin. Case Rep. 2022, 10, e05461. [Google Scholar] [CrossRef]
  20. Chan, J.F.; Chan, T.S.; Gill, H.; Lam, F.Y.; Trendell-Smith, N.J.; Sridhar, S.; Tse, H.; Lau, S.K.; Hung, I.F.; Yuen, K.Y.; et al. Disseminated Infections with Talaromyces marneffei in Non-AIDS Patients Given Monoclonal Antibodies against CD20 and Kinase Inhibitors. Emerg. Infect. Dis. 2015, 21, 1101–1106. [Google Scholar] [CrossRef]
  21. Lee, S.C.; Feenstra, J.; Georghiou, P.R. Pneumocystis jiroveci pneumonitis complicating ruxolitinib therapy. BMJ Case Rep. 2014, 2014, bcr2014204950. [Google Scholar] [CrossRef]
  22. Marples, R.; Micallef, M.; Whyte, C. Ruxolitinib-Associated Phaeohyphomycosis: A Case Report. Cureus 2021, 13, e19335. [Google Scholar] [CrossRef]
  23. Sylvine, P.; Thomas, S.; Pirayeh, E. Infections associated with ruxolitinib: Study in the French Pharmacovigilance database. Ann. Hematol. 2018, 97, 913–914. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, J.C.; Yuan, C.T.; Lin, C.C. Cutaneous cryptococcosis in a patient with myelofibrosis receiving JAK-inhibitor. EJHaem 2022, 3, 252–253. [Google Scholar] [CrossRef] [PubMed]
  25. Ho, P.J.; Bajel, A.; Burbury, K.; Dunlop, L.; Durrant, S.; Forsyth, C.; Perkins, A.C.; Ross, D.M. A case-based discussion of clinical problems in the management of patients treated with ruxolitinib for myelofibrosis. Intern. Med. J. 2017, 47, 262–268. [Google Scholar] [CrossRef] [PubMed]
  26. Kaman, N.; Ishak, A.; Muhammad, J. Unresolved chronic ulcerated nodules: Disseminated Cutaneous Sporotrichosis. Bangladesh J. Med. Sci. 2022, 21, 191–195. [Google Scholar] [CrossRef]
  27. Kasemchaiyanun, A.; Suwatanapongched, T.; Incharoen, P.; Plumworasawat, S.; Bruminhent, J. Combined Pulmonary Tuberculosis with Pulmonary and Pleural Cryptococcosis in a Patient Receiving Ruxolitinib Therapy. Infect. Drug Resist. 2021, 14, 3901–3905. [Google Scholar] [CrossRef] [PubMed]
  28. Sayabovorn, N.; Chongtrakool, P.; Chayakulkeeree, M. Cryptococcal fungemia and Mycobacterium haemophilum cellulitis in a patient receiving ruxolitinib: A case report and literature review. BMC Infect. Dis. 2021, 21, 27. [Google Scholar] [CrossRef] [PubMed]
  29. Jiang, R.; Kim, S.R.; Panse, G.; Zubek, A. Tender violaceous nodule on the palm. JAAD Case Rep. 2022, 24, 42–44. [Google Scholar] [CrossRef] [PubMed]
  30. Chakrabarti, A.; Sood, N. Cryptococcal meningitis in an immunocompetent patient with primary myelofibrosis on long-term ruxolitinib: Report of a rare case and review of literature. memo Mag. Eur. Med. Oncol. 2018, 11, 348–350. [Google Scholar] [CrossRef]
  31. Hsu, A.; Ulici, V. Verruconis gallopava in a patient with myelofibrosis on ruxolitinib. Blood 2020, 135, 1189. [Google Scholar] [CrossRef]
  32. Sugiura, H.; Nishimori, H.; Nishii, K.; Toji, T.; Fujii, K.; Fujii, N.; Matsuoka, K.I.; Nakata, K.; Kiura, K.; Maeda, Y. Secondary Pulmonary Alveolar Proteinosis Associated with Primary Myelofibrosis and Ruxolitinib Treatment: An Autopsy Case. Intern. Med. 2020, 59, 2023–2028. [Google Scholar] [CrossRef]
  33. Romani, L. Immunity to fungal infections. Nat. Rev. Immunol. 2011, 11, 275–288. [Google Scholar] [CrossRef]
  34. Reinwald, M.; Silva, J.T.; Mueller, N.J.; Fortún, J.; Garzoni, C.; de Fijter, J.W.; Fernández-Ruiz, M.; Grossi, P.; Aguado, J.M. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: An infectious diseases perspective (Intracellular signaling pathways: Tyrosine kinase and mTOR inhibitors). Clin. Microbiol. Infect. 2018, 24 (Suppl. S2), S53–S70. [Google Scholar] [CrossRef]
  35. Bechman, K.; Galloway, J.B.; Winthrop, K.L. Small-Molecule Protein Kinases Inhibitors and the Risk of Fungal Infections. Curr. Fungal Infect. Rep. 2019, 13, 229–243. [Google Scholar] [CrossRef]
  36. Harrison, C.N.; Vannucchi, A.M.; Kiladjian, J.J.; Al-Ali, H.K.; Gisslinger, H.; Knoops, L.; Cervantes, F.; Jones, M.M.; Sun, K.; McQuitty, M.; et al. Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs best available therapy for myelofibrosis. Leukemia 2016, 30, 1701–1707. [Google Scholar] [CrossRef] [PubMed]
  37. Kiladjian, J.J.; Zachee, P.; Hino, M.; Pane, F.; Masszi, T.; Harrison, C.N.; Mesa, R.; Miller, C.B.; Passamonti, F.; Durrant, S.; et al. Long-term efficacy and safety of ruxolitinib versus best available therapy in polycythaemia vera (RESPONSE): 5-year follow up of a phase 3 study. Lancet Haematol. 2020, 7, e226–e237. [Google Scholar] [CrossRef] [PubMed]
  38. Passamonti, F.; Palandri, F.; Saydam, G.; Callum, J.; Devos, T.; Guglielmelli, P.; Vannucchi, A.M.; Zor, E.; Zuurman, M.; Gilotti, G.; et al. Ruxolitinib versus best available therapy in inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): 5-year follow up of a randomised, phase 3b study. Lancet Haematol. 2022, 9, e480–e492. [Google Scholar] [CrossRef]
  39. Landtblom, A.R.; Andersson, T.M.; Dickman, P.W.; Smedby, K.E.; Eloranta, S.; Batyrbekova, N.; Samuelsson, J.; Björkholm, M.; Hultcrantz, M. Risk of infections in patients with myeloproliferative neoplasms-a population-based cohort study of 8363 patients. Leukemia 2021, 35, 476–484. [Google Scholar] [CrossRef] [PubMed]
  40. Prakash, K.; Richman, D. A case report of disseminated histoplasmosis and concurrent cryptococcal meningitis in a patient treated with ruxolitinib. BMC Infect. Dis. 2019, 19, 287. [Google Scholar] [CrossRef]
  41. Zeitler, K.; Jariwala, R.; Alrabaa, S.; Sriaroon, C. Disseminated blastomycosis in a patient with polycythemia vera on ruxolitinib. BMJ Case Rep. 2021, 14, e243694. [Google Scholar] [CrossRef]
  42. McLornan, D.P.; Khan, A.A.; Harrison, C.N. Immunological Consequences of JAK Inhibition: Friend or Foe? Curr. Hematol. Malig. Rep. 2015, 10, 370–379. [Google Scholar] [CrossRef]
  43. Heine, A.; Brossart, P.; Wolf, D. Ruxolitinib is a potent immunosuppressive compound: Is it time for anti-infective prophylaxis? Blood 2013, 122, 3843–3844. [Google Scholar] [CrossRef] [PubMed]
  44. Talpaz, M.; Kiladjian, J.J. Fedratinib, a newly approved treatment for patients with myeloproliferative neoplasm-associated myelofibrosis. Leukemia 2021, 35, 1–17. [Google Scholar] [CrossRef] [PubMed]
  45. Safdar, A.; Rodriguez, G.; Zuniga, J.; Al Akhrass, F.; Georgescu, G.; Pande, A. Granulocyte macrophage colony-stimulating factor in 66 patients with myeloid or lymphoid neoplasms and recipients of hematopoietic stem cell transplantation with invasive fungal disease. Acta Haematol. 2013, 129, 26–34. [Google Scholar] [CrossRef] [PubMed]
  46. Stevens, D.A.; Brummer, E.; Clemons, K.V. Interferon-γ as an Antifungal. J. Infect. Dis. 2006, 194, S33–S37. [Google Scholar] [CrossRef] [PubMed]
  47. Wurster, S.; Watowich, S.S.; Kontoyiannis, D.P. Checkpoint inhibitors as immunotherapy for fungal infections: Promises, challenges, and unanswered questions. Front. Immunol. 2022, 13, 1018202. [Google Scholar] [CrossRef]
  48. Kontoyiannis, D.P.; Patterson, T.F. Diagnosis and treatment of invasive fungal infections in the cancer patient: Recent progress and ongoing questions. Clin. Infect. Dis. 2014, 59 (Suppl. S5), S356–S359. [Google Scholar] [CrossRef] [PubMed]
  49. Little, J.S.; Weiss, Z.F.; Hammond, S.P. Invasive Fungal Infections and Targeted Therapies in Hematological Malignancies. J. Fungi 2021, 7, 1058. [Google Scholar] [CrossRef] [PubMed]
  50. Chamilos, G.; Lionakis, M.S.; Kontoyiannis, D.P. Call for Action: Invasive Fungal Infections Associated with Ibrutinib and Other Small Molecule Kinase Inhibitors Targeting Immune Signaling Pathways. Clin. Infect. Dis. 2018, 66, 140–148. [Google Scholar] [CrossRef] [PubMed]
  51. Mazi, P.B.; Sahrmann, J.M.; Olsen, M.A.; Coler-Reilly, A.; Rauseo, A.M.; Pullen, M.; Zuniga-Moya, J.C.; Powderly, W.G.; Spec, A. The Geographic Distribution of Dimorphic Mycoses in the United States for the Modern Era. Clin. Infect. Dis. 2023, 76, 1295–1301. [Google Scholar] [CrossRef]
  52. Taylor, M.L.; Reyes-Montes, M.D.R.; Estrada-Bárcenas, D.A.; Zancopé-Oliveira, R.M.; Rodríguez-Arellanes, G.; Ramírez, J.A. Considerations about the Geographic Distribution of Histoplasma Species. Appl. Environ. Microbiol. 2022, 88, e0201021. [Google Scholar] [CrossRef]
  53. Seidel, D.; Wurster, S.; Jenks, J.D.; Sati, H.; Gangneux, J.-P.; Egger, M.; Alastruey-Izquierdo, A.; Ford, N.P.; Chowdhary, A.; Sprute, R.; et al. Impact of climate change and natural disasters on fungal infections. Lancet Microbe 2024. online ahead of issue publication. [Google Scholar] [CrossRef]
  54. Panackal, A.A.; Hajjeh, R.A.; Cetron, M.S.; Warnock, D.W. Fungal infections among returning travelers. Clin. Infect. Dis. 2002, 35, 1088–1095. [Google Scholar] [CrossRef] [PubMed]
  55. Tenório, B.G.; Kollath, D.R.; Gade, L.; Litvintseva, A.P.; Chiller, T.; Jenness, J.S.; Stajich, J.E.; Matute, D.R.; Hanzlicek, A.S.; Barker, B.M.; et al. Tracing histoplasmosis genomic epidemiology and species occurrence across the USA. Emerg. Microbes Infect. 2024, 13, 2315960. [Google Scholar] [CrossRef] [PubMed]
  56. Gugnani, H.C.; Denning, D.W. Infection of bats with Histoplasma species. Med. Mycol. 2023, 61, myad080. [Google Scholar] [CrossRef]
  57. Sipsas, N.V.; Kontoyiannis, D.P. Occupation, Lifestyle, Diet, and Invasive Fungal Infections. Infection 2008, 36, 515–525. [Google Scholar] [CrossRef] [PubMed]
  58. Granger, D.; Streck, N.T.; Theel, E.S. Detection of Histoplasma capsulatum and Blastomyces dermatitidis antigens in serum using a single quantitative enzyme immunoassay. J. Clin. Microbiol. 2023, 62, e0121323. [Google Scholar] [CrossRef] [PubMed]
  59. Harrington, P.; Doores, K.J.; Saunders, J.; de Lord, M.; Saha, C.; Lechmere, T.; Khan, H.; Lam, H.P.J.; Reilly, A.O.; Woodley, C.; et al. Impaired humoral and T cell response to vaccination against SARS-CoV-2 in chronic myeloproliferative neoplasm patients treated with ruxolitinib. Blood Cancer J. 2022, 12, 73. [Google Scholar] [CrossRef]
  60. Egan, L.; Connolly, P.; Wheat, L.J.; Fuller, D.; Dais, T.E.; Knox, K.S.; Hage, C.A. Histoplasmosis as a cause for a positive Fungitell™ (1→3)-beta-D-glucan test. Med. Mycol. 2008, 46, 93–95. [Google Scholar] [CrossRef] [PubMed]
  61. Nguyen, M.H.; Davis, M.R.; Wittenberg, R.; McHardy, I.; Baddley, J.W.; Young, B.Y.; Odermatt, A.; Thompson, G.R. Posaconazole Serum Drug Levels Associated with Pseudohyperaldosteronism. Clin. Infect. Dis. 2020, 70, 2593–2598. [Google Scholar] [CrossRef]
  62. Dipippo, A.J.; Kontoyiannis, D.P. Azole-Associated Pseudohyperaldosteronism: A Class Effect or Azole-Specific? Clin. Infect. Dis. 2020, 71, 467–468. [Google Scholar] [CrossRef]
  63. Ji, H.H.; Tang, X.W.; Zhang, N.; Huo, B.N.; Liu, Y.; Song, L.; Jia, Y.T. Antifungal Therapy with Azoles Induced the Syndrome of Acquired Apparent Mineralocorticoid Excess: A Literature and Database Analysis. Antimicrob. Agents Chemother. 2022, 66, e0166821. [Google Scholar] [CrossRef]
  64. Wheat, L.J.; Freifeld, A.G.; Kleiman, M.B.; Baddley, J.W.; McKinsey, D.S.; Loyd, J.E.; Kauffman, C.A. Clinical Practice Guidelines for the Management of Patients with Histoplasmosis: 2007 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2007, 45, 807–825. [Google Scholar] [CrossRef] [PubMed]
  65. Saullo, J.L.; Miller, R.A. Updates on Histoplasmosis in Solid Organ Transplantation. Curr. Fungal Infect. Rep. 2022, 16, 165–178. [Google Scholar] [CrossRef] [PubMed]
  66. Brilhante, R.S.N.; Guedes, G.M.M.; Silva, M.; Castelo-Branco, D.; Cordeiro, R.A.; Sidrim, J.J.C.; Rocha, M.F.G. A proposal for antifungal epidemiological cut-off values against Histoplasma capsulatum var. capsulatum based on the susceptibility of isolates from HIV-infected patients with disseminated histoplasmosis in Northeast Brazil. Int. J. Antimicrob. Agents 2018, 52, 272–277. [Google Scholar] [CrossRef] [PubMed]
  67. Thompson, G.R., 3rd; Wiederhold, N.P. Isavuconazole: A comprehensive review of spectrum of activity of a new triazole. Mycopathologia 2010, 170, 291–313. [Google Scholar] [CrossRef] [PubMed]
  68. Lewis, J.S., 2nd; Wiederhold, N.P.; Hakki, M.; Thompson, G.R., 3rd. New Perspectives on Antimicrobial Agents: Isavuconazole. Antimicrob. Agents Chemother. 2022, 66, e0017722. [Google Scholar] [CrossRef] [PubMed]
  69. Spec, A.; Connolly, P.; Montejano, R.; Wheat, L.J. In vitro activity of isavuconazole against fluconazole-resistant isolates of Histoplasma capsulatum. Med. Mycol. 2018, 56, 834–837. [Google Scholar] [CrossRef]
  70. Moruno-Rodríguez, A.; Sánchez-Vicente, J.L.; Rueda-Rueda, T.; Lechón-Caballero, B.; Muñoz-Morales, A.; López-Herrero, F. Invasive aspergillosis manifesting as retinal necrosis in a patient treated with ruxolitinib. Arch. Soc. Esp. Oftalmol. (Engl. Ed.) 2019, 94, 237–241. [Google Scholar] [CrossRef]
Figure 1. A 59-year-old woman with myelofibrosis and disseminated histoplasmosis after 7 months of ruxolitinib. (A) Left subclavicular lymphadenopathy (yellow arrow). (B) Splenomegaly (spleen, 19 cm in craniocaudal dimension) with splenic infarction (white asterisk). (C) Histoplasma recovered from bone marrow culture (×400 magnification).
Figure 1. A 59-year-old woman with myelofibrosis and disseminated histoplasmosis after 7 months of ruxolitinib. (A) Left subclavicular lymphadenopathy (yellow arrow). (B) Splenomegaly (spleen, 19 cm in craniocaudal dimension) with splenic infarction (white asterisk). (C) Histoplasma recovered from bone marrow culture (×400 magnification).
Jof 10 00264 g001
Figure 2. Serial PET scan in our patient with disseminated histoplasmosis. (A) PET showed newly developed mediastinum lymphadenopathy (red square) while the patient was receiving intravenous liposomal amphotericin B (3 mg/kg/daily) and oral itraconazole. (B) After switching to posaconazole and increased liposomal amphotericin B (5 mg/kg/daily), follow-up PET showed decreasing F-18 FDG avidity of the mediastinal and hilar lymph nodes. Abbreviation: PET—positron emission tomography.
Figure 2. Serial PET scan in our patient with disseminated histoplasmosis. (A) PET showed newly developed mediastinum lymphadenopathy (red square) while the patient was receiving intravenous liposomal amphotericin B (3 mg/kg/daily) and oral itraconazole. (B) After switching to posaconazole and increased liposomal amphotericin B (5 mg/kg/daily), follow-up PET showed decreasing F-18 FDG avidity of the mediastinal and hilar lymph nodes. Abbreviation: PET—positron emission tomography.
Jof 10 00264 g002
Figure 3. The clinical course of the presented case. a. MDACC providers were made aware of this result. Abbreviations: +—positive; −—negative; BM Bx—bone marrow biopsy; Cx—culture; ITC—itraconazole; LAD—lymphadenopathy; LAmB 3 QD—liposomal amphotericin B 3 mg/kg/day; LAmB 5 QD—liposomal amphotericin B 5 mg/kg/day; LAmB 5 TIW—liposomal amphotericin B 5 mg/kg three times per week; LAmB 5 QW—liposomal amphotericin B 5 mg/kg weekly; MDACC—MD Anderson Cancer Center; OSH—outside hospital; PET—positron emission tomography; SAb—serum antibody; Q12H—every 12 h; QD—daily; UAg—urine antigen.
Figure 3. The clinical course of the presented case. a. MDACC providers were made aware of this result. Abbreviations: +—positive; −—negative; BM Bx—bone marrow biopsy; Cx—culture; ITC—itraconazole; LAD—lymphadenopathy; LAmB 3 QD—liposomal amphotericin B 3 mg/kg/day; LAmB 5 QD—liposomal amphotericin B 5 mg/kg/day; LAmB 5 TIW—liposomal amphotericin B 5 mg/kg three times per week; LAmB 5 QW—liposomal amphotericin B 5 mg/kg weekly; MDACC—MD Anderson Cancer Center; OSH—outside hospital; PET—positron emission tomography; SAb—serum antibody; Q12H—every 12 h; QD—daily; UAg—urine antigen.
Jof 10 00264 g003
Table 1. Incidence of invasive fungal infection in patients who received ruxolitinib.
Table 1. Incidence of invasive fungal infection in patients who received ruxolitinib.
Author, Year, Reference Study TypeType of DiseaseIncidence
Polverelli, 2016 [7,8]prospectivemyelofibrosis0.6% (3/507) a
Verstivsek, 2017 [9]clinical trialmyelofibrosis2.6% (4/155) b
Kusne, 2020 [10]retrospectiveNot available c 3.7% (5/135) d
Gold, 2022 [11]retrospectiveNot available c 4.9% (26/531) e
a. Candidiasis (1); aspergillosis (2). b. Details of fungal infections are not available. c. In these studies, ruxolitinib was used for polycythemia vera, essential thrombocythemia, myelofibrosis, and graft-vs-host disease. d. Coccidioidomycosis (5). This study specifically examined the incidence of coccidioidomycosis in patients who received ruxolitinib. e. Candidiasis (6); aspergillosis (5); Pneumocystis jiroveci pneumonia (3); coccidioidomycosis (1); cryptococcosis (1); histoplasmosis (1); not specified (9).
Table 2. Clinical presentation of 28 myelofibrosis patients on ruxolitinib with invasive fungal infection.
Table 2. Clinical presentation of 28 myelofibrosis patients on ruxolitinib with invasive fungal infection.
Patient Characteristic
Sex
 Male 18 (69%) a
Age, median year (IQR)70 (65–73) a
MF type
 Primary MF15 (54%)
 Post-PV MF5 (18%)
 Post-ET NF2 (7%)
 Unknown 6 (21%)
Associated conditions
 Splenomegaly at the onset of IFI11 (39%)
 Diabetes mellitus2 (7%)
Co-infection5 (18%) b
Time from initiation of ruxolitinib to the onset of IFI, medium months (IQR)11 (6–27) c
Ruxolitinib treatment after diagnosis of IFI d
 Discontinued permanently 13 (76%)
 Discontinued and restarted once IFI resolved3 (18%)
 Continued 1 (6%)
Antifungal prophylaxis at the onset of IFI0 (0%)
IFI, location
 Disseminated11 (39%)
 Pulmonary 6 (21%)
 Central nervous system 5 (18%)
 Other localized infection e 6 (21%)
Fungal infection, pathogen
 Cryptococcus spp.13 (46%)
 Coccidioides spp.3 (11%)
 Pneumocystis jirovecii3 (11%)
 Others f9 (32%)
All-cause mortality5 (24%) g,h
a. Data are available for 26 patients. b. One case each of Klebsiella pneumoniae bacteremia, pulmonary tuberculosis, pneumonia due to Mycobacterium avium complex, and Mycobacterium haemophilum skin infection. One patient has disseminated tuberculosis and shingles (herpes zoster). c. Data are available for 22 patients. d. Data are available for 17 patients. e. One case each of intestinal candidiasis, rhino-orbital mucormycosis, skin abscess due to Pleurostoma richardsiae, cutaneous coccidioidomycosis, cutaneous sporotrichosis, and cutaneous phaeohyphomycosis (Exophiala sp.). f. One case each is due to Aspergillus sp., Candida albicans, Exophiala sp., Histoplasma capsulatum, Mucorales, Pleurostoma richardsiae, Sporothrix schenckii, Talaromyces marneffei, and Verruconis gallopava. g. Data are available for 21 patients. h. 5 patients expired on days 7, 14, 22, 48, and 65 of hospitalization.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chiu, C.-Y.; John, T.M.; Matsuo, T.; Wurster, S.; Hicklen, R.S.; Khattak, R.R.; Ariza-Heredia, E.J.; Bose, P.; Kontoyiannis, D.P. Disseminated Histoplasmosis in a Patient with Myelofibrosis on Ruxolitinib: A Case Report and Review of the Literature on Ruxolitinib-Associated Invasive Fungal Infections. J. Fungi 2024, 10, 264. https://doi.org/10.3390/jof10040264

AMA Style

Chiu C-Y, John TM, Matsuo T, Wurster S, Hicklen RS, Khattak RR, Ariza-Heredia EJ, Bose P, Kontoyiannis DP. Disseminated Histoplasmosis in a Patient with Myelofibrosis on Ruxolitinib: A Case Report and Review of the Literature on Ruxolitinib-Associated Invasive Fungal Infections. Journal of Fungi. 2024; 10(4):264. https://doi.org/10.3390/jof10040264

Chicago/Turabian Style

Chiu, Chia-Yu, Teny M. John, Takahiro Matsuo, Sebastian Wurster, Rachel S. Hicklen, Raihaan Riaz Khattak, Ella J. Ariza-Heredia, Prithviraj Bose, and Dimitrios P. Kontoyiannis. 2024. "Disseminated Histoplasmosis in a Patient with Myelofibrosis on Ruxolitinib: A Case Report and Review of the Literature on Ruxolitinib-Associated Invasive Fungal Infections" Journal of Fungi 10, no. 4: 264. https://doi.org/10.3390/jof10040264

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop