Amjcaserep 24 E941789

You might also like

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

e-ISSN 1941-5923

© Am J Case Rep, 2023; 24: e941789


DOI: 10.12659/AJCR.941789

Received:
Accepted:
2023.07.15
2023.10.04 Immune Checkpoint Inhibitor-Induced Pure Red
Available online:
Published:
2023.10.13
2023.11.14 Cell Aplasia: A Review of 2 Cases in Metastatic
Melanoma

Authors’ Contribution: ABCDEFG 1,2 Libardo Rueda Prada 1 Mayo Clinic College of Medicine and Science, Rochester, MN, USA
Study Design A ABCDE 1,2 Tatjana Gavrancic 2 Department of Hospital Medicine, Mayo Clinic, Jacksonville, FL, USA
Data Collection B 3 Department of Internal Medicine, Universidad Industrial de Santander,
Statistical Analysis C DF 3 Miguel O. Cadena Sanabria Bucaramanga, Santander, Colombia
Data Interpretation D CDE 1,4 Igor Dumic 4 Department of Hospital Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
Manuscript Preparation E B 5 Kirk Bourgeois 5 Department of Pathology, Mayo Clinic, Jacksonville, FL, USA
Literature Search F 6 Division of Hemopathology, Mayo Clinic, Rochester, MN, USA
Funds Collection G B 6 Rebecca L. King 7 Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, USA
DEF 1,7 Yiyi Yan

Corresponding Author: Libardo Rueda Prada, e-mail: Prada.libardo@mayo.edu


Financial support: This study was supported by institutional funding from the Mayo Clinic
Conflict of interest: None declared

Case series
Patients: Female, 56-year-old • Female, 25-year-old
Final Diagnosis: Immune checkpoint inhibitor induced pure red cell aplasia
Symptoms: Anemia
Clinical Procedure: —
Specialty: Hematology • General and Internal Medicine • Oncolog

Objective: Rare disease


Background: Immunotherapy is a novel treatment offering an alternative to traditional chemotherapeutic agents for dif-
ferent malignancies. Hematologic adverse reactions (HARs) related to immune checkpoint inhibitors (ICIs) are
uncommon. Pure red cell aplasia (PRCA) is a rare hematologic complication of ICI therapy in metastatic mela-
noma with significant mortality risk despite treatment with steroids or immunosuppressive therapy. For unex-
plained acute anemia after exclusion of other causes, performing bone marrow biopsy is imperative to diag-
nose PRCA and rule out involvement of bone marrow by primary tumor. HARs can occur during ICI therapy or
even after ICI therapy is stopped. ICI rechallenge, even after the development of HARs, is considered in some
patients with good response to treatment of HARs from ICIs. Recurrence of HARs with the same or different
type of reaction is seen in some patients.
Case Reports: Two cases of ICI-induced PRCA were confirmed on bone marrow biopsy after dual ICI treatment with nivolum-
ab and ipilimumab in metastatic melanoma. In case 2, PRCA was successfully treated with steroids and later
rechallenged with single-agent nivolumab, causing mild ICI-induced immune thrombocytopenia, which did not
require treatment with steroids.
Conclusions: It is crucial to increase clinician awareness of the possibility of PRCA development not only during treatment
with ICI but also after finishing treatment with ICI; there is high mortality associated with missing an opportu-
nity to diagnose and treat PRCA on time with favorable results. ICI rechallenge can be considered in patients
who showed response to immunotherapy, especially those with limited alternative therapeutic options.

Keywords: Immune Checkpoint Inhibitors • Immunotherapy • Melanoma • Red-Cell Aplasia, Pure

Full-text PDF: https://www.amjcaserep.com/abstract/index/idArt/941789

3085   2   2   33

Publisher’s note: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated
organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be
made by its manufacturer, is not guaranteed or endorsed by the publisher

This work is licensed under Creative Common Attribution- Indexed in: [PMC] [PubMed] [Emerging Sources Citation Index (ESCI)]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e941789-1 [Web of Science by Clarivate]
Rueda Prada L. et al:
ICI-induced pure red cell aplasia in metastatic melanoma
© Am J Case Rep, 2023; 24: e941789

Background Case Reports

Pure red cell aplasia (PRCA) is a rare syndrome manifested by Case 1


marked reticulocytopenia, relative absence of erythroid pre-
cursors on bone marrow examination, and normocytic normo- A 56-year-old woman presented to the Oncology Clinic for
chromic anemia due to erythropoiesis failure [1-3]. Congenital evaluation of acute anemia. Her past medical history included
forms of PRCA are Diamond-Blackfan anemia and Pearson syn- stage IV BRAF wild-type metastatic cutaneous melanoma to the
drome, but these forms are much less common than acquired brain, lung, liver, and bones. She previously received pembro-
PRCA, which is further classified as primary or secondary [1-3]. lizumab, experiencing disease progression, and recently com-
Secondary acquired PRCA is the most common form, and its pleted the second cycle of dual immunotherapy with nivolum-
etiologies vary, with infections, autoimmune conditions, solid ab and ipilimumab 4 weeks before presentation. She had no
tumors, and medication-related adverse effects being the most history of prior gastrointestinal bleeding.
common. Multiple medications have been previously reported
to cause PRCA, and, for most of these, the exact mechanism Her initial vital signs were within normal limits. Physical exam-
causing PRCA is unclear. Commonly used medications asso- ination was significant for a pale, alert patient. The remainder
ciated with secondary PRCA include azathioprine, allopurinol, of her physical examination was within normal limits, except
carbamazepine, clopidogrel, linezolid, tacrolimus, valproic acid, for a 3-cm sacral wound with mild surrounding redness, green-
and trimethoprim-sulfamethoxazole [4-6]. Immune checkpoint ish discharge, and no bone exposure. Initial laboratory work-
inhibitors (ICIs) are promising immunotherapy agents with ex- up was remarkable for normocytic anemia, with a hemoglobin
panding indications for multiple cancers. Their association with level of 7.4 g/dL (baseline 13.1 g/dL, 3 weeks prior), chron-
PRCA is established but remains rare [7,8]. Here we report 2 ic hyponatremia, with a sodium level stable at 133 mmol/L,
cases of PRCA related to immunotherapy for metastatic mel- creatinine level of 0.97 mg/dL (at baseline), albumin level of
anoma and discuss differential diagnosis and management. 3.3 g/dL, and mild chronic aspartate transaminase and alka-
line phosphatase elevation at 61 U/L and 181 U/L, respectively.

Table 1. Case 1. Summary of laboratory workup and timeline of hospital interventions.


2 units 1 unit Bone marrow biopsy
PRBC PRBC
Interventions

Time of First 1
Day 1 Day 2 Day 3 Day 4
presentation 24 h month

Hemoglobin (g/dL) 7.4 5.8 6.4 10.8 11.3 9.8 8.6

Platelet count (×109/L) 277 270 223 222 188 235 240

White blood cell count


4.9 5.2 5.7 4.4 4.0 3.6 3.5
(×109/L)

Reticulocyte (%) 0.32 0.81 0.36 0.31 0.25 6.38

Absolute reticulocytes 9.5 31.8 14.4 12.7 9.3 208.6

Iron 85 mcg/dL. Total iron binding capacity 188 mcg/dL. TSAT 45%. Ferritin 2307 mcg/L.
Additional anemia workup B12 level >1400 ng/L. Folate level 10.0 mcg/L. Fecal occult blood test negative. Normal peripheral
blood flow cytometric immunophenotyping. Parvovirus B19 serology and PCR negative

DIC workup D-dimer 2619 ng/mL. Fibrinogen 349 mg/dL. PT 13.9 sec. INR 1.3. aPTT 29 sec

No schistocytes seen in peripheral blood smear. LDH 794 U/L (chronically elevated 624-781 U/L
Hemolysis workup
over last year). Total bilirubin 0.6 mg/dL. Haptoglobin < 14 mg/dL. Direct Coombs negative

Reference ranges: Reticulocyte (%) 0.60-2.71. Absolute reticulocyte 30.4-110.9×109/L. Iron 35-145 mcg/dL. Total iron binding capacity
250-400 mcg/dL. TSAT 14-50%. Ferritin 11-307 mcg/L. B12 level 180-914 ng/L. Folate level ³4.0 mcg/L. D-dimer £500 ng/mL.
Fibrinogen 200-393 mg/dL. PT 9.4-12.5 sec. INR 0.9-1.1. aPTT 25-37 sec. LDH 122-222 U/L. Total bilirubin £1.2 mg/dL.
Haptoglobin 30-200 mg/dL.
DIC – disseminated intravascular coagulation; LDH – lactate dehydrogenase; PRBC – packed red blood cells; PT – prothrombin time;
aPTT – activated partial thromboplastin time; TSAT – transferrin saturation.

This work is licensed under Creative Common Attribution- Indexed in: [PMC] [PubMed] [Emerging Sources Citation Index (ESCI)]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e941789-2 [Web of Science by Clarivate]
Rueda Prada L. et al:
ICI-induced pure red cell aplasia in metastatic melanoma
© Am J Case Rep, 2023; 24: e941789

A D

B E

Figure 1. Bone marrow biopsy revealed a hypercellular


C
marrow with trilineage hematopoiesis. (A)
Hematoxylin and eosin staining (magnification ×10).
Immunohistochemistry studies show (B) decreased
erythrocytosis by CD71 (×10) that are primarily
erythrocyte precursors by (C) E-cadherin (×10),
indicating minimal maturing erythroid lineage present
beyond erythrocyte precursors. No evidence of
melanoma by (D) Melan-A (×10), and (E) SOX10 (×10).

Intravenous (i.v.) ceftriaxone, vancomycin, and flagyl were ini-


tiated to cover for a possible infected sacral wound and uri-
nary tract infection. Her hemoglobin level dropped in the first
24 h, to 5.8 g/dL. Her hemoglobin level improved after a to-
The urinalysis results were suggestive of infection. Additional tal of 3 units of packed red blood cells. Her reticulocyte count
laboratory workup is summarized in Table 1. No leukopenia or was persistently low. The urine culture result was positive for
thrombocytopenia was observed. There was persistent reticu- pan-sensitive E. coli. Her sacral ulcer improved with wound
locytopenia, which improved approximately 1 month after the care measures. There were no signs of abscess. Antibiotics
ICI was stopped. Despite an elevated D-dimer level, disseminat- were deescalated to i.v. ceftriaxone, and 7 days of treatment
ed intravascular coagulation was considered less likely given were completed. The Hematology Department was consult-
a fibrinogen level >100 mg/dL, absence of thrombocytopenia, ed for suspected PRCA related to immunotherapy. Bone mar-
minimally elevated prothrombin time/international normalized row biopsy results showed slight hypercellularity (60%) with
ratio, normal activated partial prothrombin time, and absence markedly decreased erythroid precursors (<5% of total) with
of clinical signs of bleeding or thrombosis. Hemolysis seemed minimal erythroid maturation, consistent with PRCA, while
less likely given the chronic lactate dehydrogenase elevation morphology and immunohistochemistry showed no evidence
in the setting of liver metastasis, absence of hyperbilirubine- of involvement of metastatic melanoma (Figure 1). However,
mia, negative Coombs test, and absence of schistocytes on additional bone marrow studies (chromosome karyotype and
peripheral blood smear. The low haptoglobin level could have Next-GeneSequencing) were both abnormal yet inconclusive
been related to metastatic liver disease. as to the etiology from myelodysplastic syndrome/clonal cy-
topenia of undetermined significance and/or melanoma. PRCA

This work is licensed under Creative Common Attribution- Indexed in: [PMC] [PubMed] [Emerging Sources Citation Index (ESCI)]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e941789-3 [Web of Science by Clarivate]
Rueda Prada L. et al:
ICI-induced pure red cell aplasia in metastatic melanoma
© Am J Case Rep, 2023; 24: e941789

diagnosis was considered based on the bone marrow findings treatment, she developed fatigue and a right-sided facial droop
mentioned, and no other conclusive alternative etiology was and was evaluated in the Emergency Department. At that time,
found on further testing. An immunotherapy rechallenge was her hemoglobin level was 6.9 g/dL (baseline 9.3 g/dL), and she
not considered, given the rapid disease progression, poor per- received 2 units of packed red blood cells. The computed to-
formance status, and concern of anemia relapse. Her hemoglo- mography (CT) scan of the head and magnetic resonance im-
bin level remained stable, with no new transfusions required. aging (MRI) of the brain were within normal limits. Possible
She was not a candidate for further systemic therapy or clini- Bell’s palsy was diagnosed.
cal trial, given her decreased performance status. The patient
transitioned her care to hospice. During the clinic visit, her vital signs were remarkable for a
temperature of 39.3 °C. The rest of her vital signs were with-
Case 2 in normal limits. At the physical examination, she was in no
acute distress. The rest of her physical examination was with-
A 25-year-old woman presented to the Oncology Clinic for eval- in normal limits, except for an erythematous macular rash on
uation of fever and fatigue. She was diagnosed with BRAF wild- both cheeks, sparing the nasolabial folds, and neurologic ex-
type metastatic mucosal melanoma in the anus 3 months ear- amination with no signs of focalization, except for right-sided
lier and had an excision of the anal mass 10 days after initial peripheral VII cranial nerve palsy.
diagnosis. Since then, she had received frontline dual immuno-
therapy with nivolumab (1 mg/kg) and ipilimumab (3 mg/kg). Initial laboratory workup was significant for normocytic ane-
She had completed 3 cycles of immunotherapy, with the most mia, with a hemoglobin level of 7.1 g/dL, unremarkable chem-
recent one 5 days before presentation. Two days after her last istry panel, with creatinine level of 0.85 mg/dL, and normal

Table 2. Case 2. Summary of laboratory workup and timeline of hospital interventions.


2 units PRBC Intravenous 2 units Bone marrow IVIG Cyclosporine
(ER evaluation) Solu-Medrol PRBC biopsy Prednisone
Interventions

Time of First 1
Day 1 Day 2 Day 3 Day 4
presentation 24 h month

Hemoglobin (g/dL) 7.1 7.2 5.4 6.9 9.5 8.6 12.8

Platelet count (×109/L) 134 140 93 102 134 119 173

White blood cell count


5.1 4.3 1.7 5.0 8.6 7.4 13.2
(×109/L)

Reticulocyte (%) <0.31 0.39 <0.31 <0.31 <0.31 <0.31 3.10

Absolute reticulocytes 8.8 11.1 8.8 8.8 8.8 8.8 354

Two months before this admission: Iron 18 mcg/dL. Total iron binding capacity 297 mcg/dL.
TSAT 6%. Ferritin 14. B12 level 320 ng/L. Folate level 7.3 mcg/L. Iron studies during this
Anemia workup​
admission: Iron 2-3 mcg/dL. Total iron binding capacity 192 mcg/dL. TSAT >90%. Ferritin 642.
Folate level and B12 level was not repeated

DIC workup​ D-dimer 555 ng/mL. Fibrinogen 111 mg/dL. PT 11.6 sec. INR 1.1. aPTT 32 sec

No schistocytes on peripheral blood smear. LDH 877 U/L. Total bilirubin 1.7 mg/dL
Hemolysis workup​ (indirect 1.3 mg/dL). Haptoglobin < 14 mg/dL. Direct Coombs negative. G6PD normal.
ANA normal. Paroxysmal nocturnal hemoglobinuria antigen normal​
Parvovirus B19 serology and PCR negative. Hepatitis A, B, C serology was negative for active
Infectious workup​
infection. HSV 1 and 2 PCR negative. HIV antigen and antibody screen negative

Reference ranges: Reticulocyte (%) 0.60-2.71. Absolute reticulocyte 30.4-110.9×109/L. Iron 35-145 mcg/dL. Total iron binding capacity
250-400 mcg/dL. TSAT 14-50%. Ferritin 11-307 mcg/L. B12 level 180-914 ng/L. Folate level ³4.0 mcg/L. D-dimer £500 ng/mL.
Fibrinogen 200-393 mg/dL. PT 9.4-12.5 sec. INR 0.9-1.1. aPTT 25-37 sec. LDH 122-222 U/L. Total bilirubin £1.2 mg/dL.
Haptoglobin 30-200 mg/dL.
ANA – antinuclear antibody; DIC – disseminated intravascular coagulation; LDH – lactate dehydrogenase; HSV – herpes simplex virus;
PRBC – packed red blood cells; PT – prothrombin time; aPTT – activated partial thromboplastin time; TSAT – transferrin saturation.

This work is licensed under Creative Common Attribution- Indexed in: [PMC] [PubMed] [Emerging Sources Citation Index (ESCI)]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e941789-4 [Web of Science by Clarivate]
Rueda Prada L. et al:
ICI-induced pure red cell aplasia in metastatic melanoma
© Am J Case Rep, 2023; 24: e941789

A B

Figure 2. (A) Bone marrow aspirate and (B) core biopsy show hypercellular bone marrow with markedly decreased and left shifted
erythropoiesis. Only rare scattered pronormoblasts were identified on the aspirate, and the biopsy showed inconspicuous
erythroid islands with virtually no maturation. There was background megakaryocytic and granulocytic hyperplasia.
(A) Wright Giemsa, 600× oil. (B) Hematoxylin and eosin, 200×.

liver enzyme and alkaline phosphatase levels. The remaining Four months later, while being off treatment, she was noticed
laboratory results are summarized in Table 2. CT of the chest/ to have new multifocal lytic bone lesions and a new lung nod-
abdomen/pelvis with i.v. contrast showed progression of the ule, so she was re-started on single-agent nivolumab. After 2
metastatic lymphadenopathy in the pelvis and progressive en- cycles, she developed acute thrombocytopenia (platelet count
largement of a metastatic lesion in the right ilium. Blood cul- of 66×109/L, which resolved after a course of prednisone), sug-
ture results were negative. Antibiotics were not initiated, ow- gestive of mild immune thrombocytopenia secondary to ICI;
ing to a low suspicion for infection. Disseminated intravascular however, her hemoglobin level remained stable, and therefore
coagulation was considered less likely. Hemolysis was consid- treatment was continued. Four months after having been on
ered in the differential diagnosis, based on additional test re- nivolumab, MRI of the femur suggested new metastatic dis-
sults (see Table 2). ease, and nivolumab was stopped. Carboplatin, paclitaxel, and
pembrolizumab were then initiated as salvage therapy. She
The patient was treated with i.v. Solu-Medrol 1 g daily for 5 had a partial response after 3 cycles of salvage chemo-immu-
days for probable immunotherapy-induced PRCA, with a com- notherapy. Unfortunately, she experienced thrombocytopenia
ponent of possible autoimmune hemolytic anemia. Her he- and therefore continued treatment with carboplatin, reduced
moglobin level dropped in the first day to 5.4 g/dL. She re- to AUC 4. After 6 cycles, the patient had a positive response
ceived a transfusion with 2 units of packed red blood cells. with no new metastatic disease and was placed on a treatment
Her hemoglobin level improved to 6.9 g/dL on day 2 and to holiday per her preference. Chemo-immunotherapy was rein-
9.5 g/dL on day 3, without additional transfusion needed. A itiated 3 months later because of symptomatic disease pro-
bone marrow aspirate and biopsy (Figure 2) showed a slight- gression. She was able to tolerate the treatment for the next
ly hypercellular marrow for age (90%), with decreased and 6 months, until she was found to have cerebral metastatic dis-
markedly left-shifted erythropoiesis and increased granulo- ease, for which she received radiation therapy, followed by ad-
poiesis and megakaryocytes. There was marked anisopoi- missions for rectal bleeding and lumbar spine pathologic frac-
kilocytosis with occasional microcytic cells and slight ellipto- ture, causing cauda equina. She died 6 months later.
cytes, white blood cells without cytologic abnormalities, and
platelets without cytologic abnormalities. The myeloid: ery-
throid ratio was >10: 1. Erythroid precursors quantity was de- Discussion
creased, left shifted maturation with predominantly pronor-
moblasts. Myeloid precursors quantity was increased, normal Our immune system plays an important role in the battle against
maturation. Iron staining was done, showing storage iron was cancer. Immune checkpoints are crucial self-tolerance path-
normal, sideroblasts were absent, and ring sideroblasts were ways that prevent the immune system from attacking healthy
not seen. There was no involvement by metastatic melano- cells. Cancer cells downregulate the immune system by multi-
ma. Parvovirus B19 staining was negative. Solu-Medrol i.v. ple mechanisms, including production of immunosuppressive
was followed by 5 days of i.v. immune globulin (IVIG), cyclo- cytokines that stimulate inhibitory immune checkpoints [9].
sporine 100 mg twice daily, and prednisone. Ipilimumab and Immunotherapy is a revolutionary therapy that enhances a
nivolumab were discontinued. patient’s immune system to destroy tumor cells. With the

This work is licensed under Creative Common Attribution- Indexed in: [PMC] [PubMed] [Emerging Sources Citation Index (ESCI)]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e941789-5 [Web of Science by Clarivate]
Rueda Prada L. et al:
ICI-induced pure red cell aplasia in metastatic melanoma
© Am J Case Rep, 2023; 24: e941789

introduction of ipilimumab in 2011, followed by pembrolizum- congenital or acquired. Acquired PRCA can be caused by au-
ab in 2014, ICIs have transformed cancer management, offering toimmune diseases, neoplasms invading bone marrow, medi-
a usually more tolerable alternative to the systemic cytotox- cations, such as azathioprine and procainamide, or can be re-
ic therapy. The use of ICIs has shown benefits in the treat- lated to infections, such as parvovirus B19.
ment of different types of malignancies, including melanoma,
Hodgkin’s lymphoma, and lung, bladder, and renal cancer [10]. Our patients did not have a history of a pre-existing autoim-
mune disease, which if present and active may have increased
Unfortunately, immunotherapy can cause immune-related their risk of developing severe irAEs with the use of ICIs. A
adverse events (irAEs) and compromise almost any organ or multicenter study of a small cohort of patients with meta-
system. The skin, digestive, pulmonary, renal, and endocrine static melanoma treated with ipilimumab who had a pre-ex-
systems are the most affected [11]. Hematologic adverse reac- isting autoimmune diseases showed a higher incidence of
tions (HARs) to immunotherapy are not common and include irAEs and autoimmune disorder flare-up [22]. These concerns
thrombocytopenia, pancytopenia or immune aplastic anemia, led to exclude patients with autoimmune diseases from clin-
neutropenia, hemolytic anemia, cytokine release syndrome ical trials. Further studies were done to clarify the risk-bene-
with hemophagocytic syndrome, bicytopenia, and PRCA [12]. fit ratio of ICIs in patients with cancer and pre-existing auto-
Immune-mediated thrombocytopenia and hemolytic anemia immune diseases. These studies indicated that the use of ICIs
are among the most common HARs seen, usually involving the may be safe in this patient population and is not an absolute
use of the ICIs nivolumab and pembrolizumab [13]. Ipilimumab indication to discontinue ICI therapy. However, there may be
has shown the shortest time from therapy initiation to on- still a risk of severe and fatal disease [23,24]. A more recent
set of HARs [13]. It is important to keep monitoring blood cell study showed similar efficacy and safety with ICI use in pa-
counts even after discontinuation of treatment, given that tients with cancer without and with pre-existing autoimmune
HAR recurrence has been seen in approximately 8.9% of cas- disease. However, impaired T-cell activation in patients with
es [13]. The mortality from HARs related to ICIs is variable and active pre-existing autoimmune disease predisposes them to
dependent on the type of disturbance, with the highest seen worse outcomes during ICI therapy. On the contrary, the de-
being aplastic anemia at 54.5%, and average mortality for all velopment of immune irAEs in patients with autoimmune dis-
adverse reactions between 12% and 15.5% [13,14]. Hence, it ease, which was found to be usually mild, may be a sign of
is important to increase clinician awareness of this condition T-cell activation, which is associated with superior immuno-
to provide prompt treatment and avoid significant mortality. therapy efficacy and better outcomes [25].

There are reports of nivolumab-induced autoimmune hemolyt- Neurological irAEs are seen in <1% of patients on ICIs [26]. Bell’s
ic anemia [15,16], some which are associated with PRCA [17]. palsy, as seen in case 2, has been rarely reported in patients on
Rare cases of PRCA have been reported without hemolysis, immunotherapy [26-28]. A single-center retrospective study of
such as the case reported by Yuki et al of a patient with car- 353 patients treated with immunotherapy between 2015 and
diac metastatic melanoma who developed PRCA at 21 months 2018 found 5 cases of Bell’s palsy, from which 3 cases were
of being on nivolumab [18]. There are several case reports of related to dual immunotherapy in patients with metastatic
aplastic anemia in patients with metastatic melanoma and oth- melanoma [27]. In some cases, it is difficult to differentiate a
er malignancies treated with dual immunotherapy [13,19-21]. paraneoplastic syndrome manifestation related to underlying
However, cases of PRCA with dual immunotherapy in patients malignancy from neurological irAEs secondary to ICIs.
with metastatic melanoma have been rarely reported. A case
series review of 15 cases of new-onset PRCA related to immu- Anemia observed in oncologic patients can be multifactorial.
notherapy from the Food and Drug Administration’s Adverse Hence, bone marrow biopsy is important to confirm the diag-
Event Reporting System from 2011 to 2022 reported 9 cases nosis of PRCA and is critical to rule out direct tumor involve-
related to metastatic melanoma, among which 6 were relat- ment of the bone marrow to determine the potential cause
ed to single immunotherapy either with pembrolizumab, ipili- of PRCA and to guide further treatment. In case 1, infection
mumab or nivolumab, and only 3 were related to dual immu- was unlikely to be the major cause for the profound anemia,
notherapy with ipilimumab and nivolumab [7]. since the laboratory findings were not compatible with infec-
tious hemolytic anemia and there were no signs of severe sys-
PRCA is rare and usually presents as a normocytic, normochro- temic infection. In addition, our patients were not on other
mic anemia with a persistent low reticulocyte count and sig- medications that could have explained their anemia, and oth-
nificant decreased or absent red blood cell precursors in bone er etiologies, such as disseminated intravascular coagulation,
marrow (< 1% erythroblasts or few proerythroblasts and/or and infections, such as parvovirus B19 infection, were ruled
basophilic erythroblasts not exceeding 5% of the marrow dif- out. Hemolysis was considered unlikely in case 1 but likely in
ferential count). Iron stains are usually normal. PRCA can be case 2, based on hemolysis workup. Although haptoglobin is

This work is licensed under Creative Common Attribution- Indexed in: [PMC] [PubMed] [Emerging Sources Citation Index (ESCI)]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e941789-6 [Web of Science by Clarivate]
Rueda Prada L. et al:
ICI-induced pure red cell aplasia in metastatic melanoma
© Am J Case Rep, 2023; 24: e941789

commonly used as one of the markers for hemolysis, its val- HARs may require additional treatments, such as IVIG, ritux-
ue should be interpreted in conjunction with other hemato- imab, or infliximab, depending on the type of HAR shown. ICI
logic evaluations to establish the diagnosis of hemolysis. In rechallenge is considered when indicated after a patient has
addition, an abnormal haptoglobin level can be seen in oth- recovered. This represents a risk for HAR recurrence, which has
er conditions, such as metastatic carcinomas or cirrhosis, in been seen in between 33% and 43% of cases [13,33]. However,
the absence of hemolysis [29]. Low haptoglobin in case 1 was not all cases have shown recurrence of HAR with the rechal-
considered possibly related to liver dysfunction from metastat- lenge of ICI therapy [13]. In addition, patients should be close-
ic disease. The patient in case 1 had chronic elevated lactate ly monitored, as they are at risk of developing another type of
dehydrogenase possible secondary to metastatic disease and HAR, as was seen in case 2, in which the patient did not have
had no acute change during this hospital admission. recurrent PRCA but did develop acute thrombocytopenia with
the immunotherapy rechallenge. Additional studies are need-
The patient in case 1 did not require any additional interven- ed to identify the risk factors that can predispose patients to
tion more than supportive blood transfusions. But in other cas- develop HAR recurrence after an ICI rechallenge. It is not well
es, such as case 2, treatment with high-dose steroid, IVIG, and known if this recurrence can be related to the long half-life of
chronic immunosuppressant is required, and it is important to these antibodies or prolonged receptor T-cell occupancy. Most
consider rechallenge with immunotherapy after clinical recovery. cases of ICI-induced PRCA had fatal outcomes, despite steroid
Final management decisions are individualized and made based or immunosuppressive treatment [7], unlike case 2.
on initial workup, severity, and clinical course, despite the initial
similar diagnosis of PRCA. Other types of HARs could be seen
with ICI rechallenge, such as the one seen in case 2, in which the Conclusions
patient developed mild immune thrombocytopenia with a single
immunotherapy rechallenge after having recovered from PRCA. Immunotherapy is a novel and revolutionary treatment being
widely used for different types of malignancies. There is a sig-
The exact mechanism of PRCA related to immunotherapy is nificant risk of irAEs with the use of immunotherapy. HARs to
unknown, but like other irAEs, it is presumed to be related to immunotherapy are not common and carry a significant mor-
widespread activation of cytotoxic T cells, production of anti- tality risk. PRCA secondary to immunotherapy in metastatic
red blood cell (or erythroid precursors) antibodies, and direct melanoma is rare and carries significant mortality risk, despite
damage to erythroblasts [30]. In fact, activated CD8+ lympho- treatment with steroids or immunosuppressive therapy. Bone
cytes seen in bone marrow specimens of patients with PRCA marrow biopsy is crucial for diagnosis and for performing ad-
support this theory [31]. Other studies have suggested a pos- ditional studies to rule out other etiologies, such as direct tu-
sible production of red blood cell antibodies against erythroid mor invasion to the bone marrow. Patients with PRCA relat-
precursors or erythropoietin, especially with ipilimumab [16,32]. ed to immunotherapy are at risk of developing other types of
Because of the rarity of PRCA related to immunotherapy, it is HARs. It is important to increase clinician awareness of this
unclear whether PRCA is more likely to occur in certain patient risk in order to provide prompt initial treatment and to con-
populations, such as according to sex, age, and other comor- tinue monitoring patients for any other HARs during immu-
bidities. In addition, the immunotherapy regimens used that notherapy rechallenge, particularly in patients who are candi-
are associated with PRCA also need to be evaluated in further dates and have no other options for treatment.
studies, especially given the introduction of a newer genera-
tion of ICIs over the past few years. Declaration of Figures’ Authenticity

Treatment of PRCA related to immunotherapy includes cortico- All figures submitted have been created by the authors who
steroids, supportive management with transfusions as need- confirm that the images are original with no duplication and
ed, and the addition of cyclosporine in refractory cases. Other have not been previously published in whole or in part.

References:
1. Means RT Jr. Pure red cell aplasia. Blood. 2016;128(21):2504-9 5. Koduri PR, Vanajakshi S, Anuradha R. Azathioprine-associated pure red cell
2. Gurnari C, Maciejewski JP. How I manage acquired pure red cell aplasia in aplasia in renal transplant recipients: A report of two cases. Ann Hematol.
adults. Blood. 2021;137(15):2001-9 2014;93(2):329-30
3. Sawada K, Hirokawa M, Fujishima N. Diagnosis and management of acquired 6. Yang XY, Chen L, Gu JN, et al. Linezolid-induced pure red cell aplasia: A case
pure red cell aplasia. Hematol Oncol Clin North Am. 2009;23(2):249-59 report. Infect Drug Resist. 2022;15:3847-56
4. Chao SC, Yang CC, Lee JY. Hypersensitivity syndrome and pure red cell apla- 7. Guo Q, Gao J, Guo H, et al. Immune checkpoint inhibitor-induced pure red
sia following allopurinol therapy in a patient with chronic kidney disease. cell aplasia: Case series and large-scale pharmacovigilance analysis. Int
Ann Pharmacother. 2005;39(9):1552-56 Immunopharmacol. 2023;114:109490

This work is licensed under Creative Common Attribution- Indexed in: [PMC] [PubMed] [Emerging Sources Citation Index (ESCI)]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e941789-7 [Web of Science by Clarivate]
Rueda Prada L. et al:
ICI-induced pure red cell aplasia in metastatic melanoma
© Am J Case Rep, 2023; 24: e941789

8. Saliba AN, Xie Z, Higgins AS, et al. Immune-related hematologic adverse 21. Younan RG, Raad RA, Sawan BY, Said R. Aplastic anemia secondary to dual
events in the context of immune checkpoint inhibitor therapy. Am J Hematol. cancer immunotherapies a physician nightmare: Case report and literature
2021;96(10):E362-E67 review. Allergy Asthma Clin Immunol. 2021;17(1):112
9. Vinay DS, Ryan EP, Pawelec G, et al. Immune evasion in cancer: Mechanistic ba- 22. Johnson DB, Sullivan RJ, Ott PA, et al. Ipilimumab therapy in patients with
sis and therapeutic strategies. Semin Cancer Biol. 2015;35(Suppl.):S185-S98 advanced melanoma and preexisting autoimmune disorders. JAMA Oncol.
10. Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune check- 2016;2(2):234-40
point inhibitors and beyond. J Hematol Oncol. 2021;14(1):45 23. Abdel-Wahab N, Shah M, Lopez-Olivo MA, Suarez-Almazor ME. Use of im-
11. Brahmer JR, Lacchetti C, Schneider BJ, et al. Management of immune-relat- mune checkpoint inhibitors in the treatment of patients with cancer and
ed adverse events in patients treated with immune checkpoint inhibitor preexisting autoimmune disease: A systematic review. Ann Intern Med.
therapy: American Society of Clinical Oncology Clinical Practice Guideline. 2018;168(2):121-30
J Clin Oncol. 2018;36(17):1714-68 24. Haanen J, Ernstoff MS, Wang Y, et al. Autoimmune diseases and immune-
12. Michot JM, Lazarovici J, Tieu A, et al. Haematological immune-related ad- checkpoint inhibitors for cancer therapy: Review of the literature and per-
verse events with immune checkpoint inhibitors, how to manage? Eur J sonalized risk-based prevention strategy. Ann Oncol. 2020;31(6):724-44
Cancer. 2019;122:72-90 25. Han CY, Fitzgerald C, Lee M, et al. Association between toxic effects and sur-
13. Ghanem P, Marrone K, Shanbhag S, et al. Current challenges of hemato- vival in patients with cancer and autoimmune disease treated with check-
logic complications due to immune checkpoint blockade: A comprehensive point inhibitor immunotherapy. JAMA Oncol. 2022;8(9):1352-54
review. Ann Hematol. 2022;101(1):1-10 26. Kichloo A, Albosta MS, Jamal SM, et al. Atezolizumab-induced Bell’s palsy
14. Davis EJ, Salem JE, Young A, et al. Hematologic complications of immune in a patient with small cell lung cancer. J Investig Med High Impact Case
checkpoint inhibitors. Oncologist. 2019;24(5):584-88 Rep. 2020;8:2324709620965010
15. Palla AR, Kennedy D, Mosharraf H, Doll D. Autoimmune hemolytic anemia 27. Yuen C, Reid P, Zhang Z, et al. Facial palsy induced by checkpoint blockade:
as a complication of nivolumab therapy. Case Rep Oncol. 2016;9(3):691-97 A single center retrospective study. J Immunother. 2019;42(3):94-96
16. Schwab KS, Heine A, Weimann T, et al. Development of hemolytic anemia in 28. Takemura K, Yamanaka T, Hayashida M, et al. Bell’s palsy during rechal-
a nivolumab-treated patient with refractory metastatic squamous cell skin lenge of immune checkpoint inhibitor. IJU Case Rep. 2023;6(2):144-46
cancer and chronic lymphatic leukemia. Case Rep Oncol. 2016;9(2):373-78 29. Shih AW, McFarlane A, Verhovsek M. Haptoglobin testing in hemolysis:
17. Nair R, Gheith S, Nair SG. Immunotherapy-associated hemolytic anemia Measurement and interpretation. Am J Hematol. 2014;89(4):443-47
with pure red-cell aplasia. N Engl J Med. 2016;374(11):1096-97 30. Young NS, Abkowitz JL, Luzzatto L. New insights into the pathophysiolo-
18. Yuki A, Takenouchi T, Takatsuka S, Ishiguro T. A case of pure red cell apla- gy of acquired cytopenias. Hematology Am Soc Hematol Educ Program.
sia during nivolumab therapy for cardiac metastatic melanoma. Melanoma 2000;18-38
Res. 2017;27(6):635-37 31. Zhuang J, Du J, Guo X, et al. Clinical diagnosis and treatment recommen-
19. Helgadottir H, Kis L, Ljungman P, et al. Lethal aplastic anemia caused by dations for immune checkpoint inhibitor-related hematological adverse
dual immune checkpoint blockade in metastatic melanoma. Ann Oncol. events. Thorac Cancer. 2020;11(3):799-804
2017;28(7):1672-73 32. Simeone E, Grimaldi AM, Esposito A, et al. Serious haematological toxici-
20. Meyers DE, Hill WF, Suo A, et al. Aplastic anemia secondary to nivolumab ty during and after ipilimumab treatment: A case series. J Med Case Rep.
and ipilimumab in a patient with metastatic melanoma: A case report. Exp 2014;8:240
Hematol Oncol. 2018;7:6 33. Delanoy N, Michot JM, Comont T, et al. Haematological immune-related
adverse events induced by anti-PD-1 or anti-PD-L1 immunotherapy: A de-
scriptive observational study. Lancet Haematol. 2019;6(1):e48-e57

This work is licensed under Creative Common Attribution- Indexed in: [PMC] [PubMed] [Emerging Sources Citation Index (ESCI)]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e941789-8 [Web of Science by Clarivate]

You might also like