TO THE EDITOR:

Vitamin C (Vc) deficiency is characterized by an array of mucocutaneous findings, including ecchymoses, petechiae, perifollicular hemorrhage, oral ulcerations, gingival hypertrophy, and corkscrew hairs. In addition, it can cause cardiovascular, pulmonary, and neuropsychiatric effects and may be fatal if left untreated.1,2 Also known as ascorbic acid, Vc acts as an essential cofactor to enzymes prolyl hydroxylase and lysyl hydroxylase, which crosslink propeptides to stabilize the structure of collagen. Scurvy, the clinical disease caused by Vc deficiency, is primarily seen in patients with restrictive diets, resource limitations, substance abuse, and malabsorption syndromes.2 Currently defined as Vc concentrations <11 μmol/L, many consider hypovitaminosis C to be represented by serum concentrations of <30 μmol/L.1 

Patients with leukemia have long been recognized to have decreased serum Vc levels,3-5 and several studies have indicated decreased survival for individuals with malignancies who are Vc deficient.6 However, there is uncertainty whether decreased serum Vc in these patients is due to increased Vc use, decreased intake, altered Vc cellular internalization mechanisms, or a combination of these factors.4,7,8 The role that Vc plays in the oncogenic pathways leading to leukemia is an area of active research. Therapeutic benefits of Vc supplementation have been investigated but, because of study limitations, remain controversial among clinical oncologists.9,10 

Herein, we identify 6 patients with acute myeloid leukemia (AML) and scurvy, diagnosed by the University of Pennsylvania Dermatology Consult Service. We postulate that because the mucocutaneous manifestations of scurvy appear similar to common sequelae of leukemia (eg, thrombocytopenia and chemotherapy-associated mucositis), signs of Vc deficiency may be inadvertently attributed to the malignancy itself or to therapeutic interventions, leading to diagnostic error. Recognition of Vc deficiency in this patient population represents an important practice gap.

In this series, we identified 4 female patients and 1 male patient with Vc deficiency (Table 1). One additional male patient (patient 6) was included in our series; he presented with findings typical of scurvy but had a Vc serum concentration of 26 μmol/L 1 week after initiating Vc supplementation. Of note, 5 patients were considered to have severe malnutrition, and half of them were on parenteral nutrition at the time of scurvy diagnosis. The parenteral nutrition formulation contained 10 mL of multivitamin injection–adult, which contains 200 mg of Vc. Patient 1 received oral supplements that contained at least 25 mg of Vc daily in addition to her hospital neutropenic diet.

Table 1.

Demographic characteristics, leukemia genetics, and therapeutic interventions of patients with low Vitamin C

Patient no.SexAge, yAML genetics Recent or current chemotherapy treatmentTreatment linePlatelet countVc level (μmol/L)Malnutrition present (yes/no) Parental nutrition
SubtypePathologic variants presentKaryotype
57 AML-MRC BRAF, NF1, NPM1, DNMT3A  46,XX,-18,+r[5]/46.XX[15] Liposomal daunorubicin-cytarabine Induction 20 Yes No 
73 M5 DNMT3A, FLT3, NPM1, BRCA2, SMC1A  47,XY,+mar[2]/46,XY[18] 7 + 3 and midostaurin Induction <5 Yes Yes 
66 M5 DNMT3A, IDH2, PTPN11, EZH2  Normal Decitabine and venetoclax R/R 24 Yes Yes 
60 t-AML IKZF1, BRAF, ETV6, RUNX1  47,XX,+8[20] Liposomal daunorubicin-cytarabine R/R 10 <5 No No 
65 t-AML NF1, TP3, CD79a  Complex (monosomal) Busulfan & cyclophosphamide Conditioning for HSCT 53 Yes Yes 
62 t-AML DNMT3A, JAK2, PLCG1  45,X,-Y,t(7;21;8)(q11.2;q22;q21.3)[19]/46,XY[1] Azacitidine and venetoclax R/R 26 Yes No 
Patient no.SexAge, yAML genetics Recent or current chemotherapy treatmentTreatment linePlatelet countVc level (μmol/L)Malnutrition present (yes/no) Parental nutrition
SubtypePathologic variants presentKaryotype
57 AML-MRC BRAF, NF1, NPM1, DNMT3A  46,XX,-18,+r[5]/46.XX[15] Liposomal daunorubicin-cytarabine Induction 20 Yes No 
73 M5 DNMT3A, FLT3, NPM1, BRCA2, SMC1A  47,XY,+mar[2]/46,XY[18] 7 + 3 and midostaurin Induction <5 Yes Yes 
66 M5 DNMT3A, IDH2, PTPN11, EZH2  Normal Decitabine and venetoclax R/R 24 Yes Yes 
60 t-AML IKZF1, BRAF, ETV6, RUNX1  47,XX,+8[20] Liposomal daunorubicin-cytarabine R/R 10 <5 No No 
65 t-AML NF1, TP3, CD79a  Complex (monosomal) Busulfan & cyclophosphamide Conditioning for HSCT 53 Yes Yes 
62 t-AML DNMT3A, JAK2, PLCG1  45,X,-Y,t(7;21;8)(q11.2;q22;q21.3)[19]/46,XY[1] Azacitidine and venetoclax R/R 26 Yes No 

AML-MRC, AML with myelodysplasia-related changes; F, female; HSCT, hematopoietic stem cell transplantation (both allo/auto); IDG2, isocitrate dehydrogenase 2; M, male; R/R, relapsed/refractory; 7 + 3, cytarabine and daunorubicin; t-AML, treatment-related AML.

At the time of AML diagnosis.

Severe protein calorie malnutrition, as established by a registered dietician.

Variant of unknown significance; however, the allele frequency was >15%.

All patients identified had AML: 3 patients had treatment-related AML, 2 had de novo AML, and 1 had AML that developed from prior myelodysplasia (see Table 1 for specific cytogenetic information). At the time of scurvy diagnosis, 3 patients were receiving cytarabine- and daunorubicin-based treatments, 2 were receiving hypomethylating agents plus venetoclax, and 1 was receiving conditioning chemotherapy for an upcoming hematopoietic stem cell transplantation. All patients had either petechiae or ecchymoses; 4 patients had mucosal ulceration or gingival bleeding, and 2 patients had the classic findings of corkscrew hairs with perifollicular hemorrhage (Figure 1). All patients had thrombocytopenia, although none were refractory to platelet transfusions (Table 1).

Figure 1.

Cutaneous manifestations of scurvy. (A) Perifollicular hemorrhage (yellow arrow) and petechiae in patient 1; Corkscrew hair in patient 1 (white arrow). (B) Petechiae and ecchymoses (yellow arrow) in patient 2. (C) Mucosal ulceration and gingival bleeding in patient 4. (D) Resolution of mucosal ulceration and gingival bleeding in patient 4 after Vc supplementation.

Figure 1.

Cutaneous manifestations of scurvy. (A) Perifollicular hemorrhage (yellow arrow) and petechiae in patient 1; Corkscrew hair in patient 1 (white arrow). (B) Petechiae and ecchymoses (yellow arrow) in patient 2. (C) Mucosal ulceration and gingival bleeding in patient 4. (D) Resolution of mucosal ulceration and gingival bleeding in patient 4 after Vc supplementation.

Close modal

Upon establishing Vc deficiency, patients 1 and 2 received between 200 and 250 mg of ascorbic acid daily. Patients 3, 4, and 6 received 1 g of ascorbic acid daily for at least 1 week. The clinical status of patient 5 deteriorated because of idiopathic pulmonary syndrome, and she died shortly after receiving her Vc serum results. All patients who received Vc repletion had rapid improvement of their ecchymoses and petechiae. Three patients had complete resolution of their mucositis, 2 of whom had resolution before absolute neutrophil count recovery. One patient (patient 3) had mucositis improvement but continued oral ulceration because of acyclovir-resistant herpes simplex virus infection.

This case series highlights the importance of recognizing the signs and symptoms of Vc deficiency, especially because these clinical clues may be misattributed to a patient’s hematologic malignancy, chemotherapy, or stem cell therapy. For all patients presented, the Dermatology department was consulted because of concern for various alternative diagnoses. As such, we encourage oncologists and dermatologists to consider the diagnosis of scurvy when evaluating these patients and replete Vc to avoid associated morbidity.

It is notable that in our institutional experience, most cases of scurvy seen in patients with hematologic malignancy have been in patients with AML. We cannot rule out a referral bias or a high prevalence of AML in our patient population. There are multiple reports of Vc deficiency in patients with non-AML hematologic malignancies, although many studies simply show lower serum Vc levels in patients with leukemia as compared with those in healthy controls.3,4,7 A larger study of patients with leukemia is required to ascertain whether AML places patients at increased risk for the development of scurvy.

We initially assumed that our patients receiving parenteral nutrition or oral supplements were at low risk for the development of scurvy. However, it may be that the recommended dietary allowance should be higher for those with hematologic malignancy, as it is for woman who are pregnant or lactating and for smokers. To our knowledge, there is no evidence to indicate decreased Vc absorption or increased Vc excretion in hematologic malignancies.11,7 Patients receiving treatment for AML often experience significant fatigue and nausea, which lead to limited food consumption and reduced Vc intake. Furthermore, complications such as chemotherapy-induced mucositis or oral infections can cause odynophagia, which may limit ingestion of acidic fruits and vegetables in particular. Although dietary recommendations have changed for patients undergoing chemotherapy and stem cell therapy, many patients still adhere to diets that warn against rough-textured fruit, preprepared salads, and precut fruits during neutropenia.12 

Various theories have been proposed regarding lower serum concentrations of Vc in patients with leukemia. Because leukocytes have high intracellular Vc concentrations, cancer-induced antioxidant depletion or chemotherapy-induced oxidative stress may lead to lower levels of Vc in patients with leukemia.7,13 It has also been established that at supraphysiologic levels, Vc can act as a pro-oxidant and selectively target malignant cells, thereby raising the question about a possible protective effect that treatment with Vc may have.11,14,15 Further work is needed to understand the role of Vc in leukemia, specifically AML, and whether there is a survival benefit of Vc supplementation in this population. However, multiple studies have shown improved quality of life in patients receiving Vc supplementation, noting few risks of therapy.9,16 Given that testing for Vc deficiency is relatively inexpensive and supplementation benign, we recommend that clinical oncologists and dermatologists maintain a low threshold for checking and repleting Vc levels in patients with AML and other hematologic malignancies.

Interestingly, a growing body of evidence shows that Vc has a unique modulatory effect on leukemic myeloid progenitor cells and may have a role in leukemogenesis.11,14,15,17,18 Vc induces apoptosis via the oxidation of reduced glutathione and activation of the Raf1 and extracellular signal-regulated kinase pathways in AML cells.14,15 As a cofactor for the ten-eleven translocation (TET) enzymes, Vc promotes DNA demethylation and dioxygenase activity, particularly notable because inactivating TET2 mutations are recurrent in AML.11,18 Although loss-of-function mutations in TET2 are common in AML, TET2 activity can also be decreased via competitive inhibition by oncometabolite 2-hydroxyglutarate, which accumulates in patients with AML-associated mutations in isocitrate dehydrogenase 1 or 2.19 It is theorized that either loss-of-function or reduced dioxygenase activity of TET2 may drive compensatory mechanisms to upregulate other TET enzyme activity and thereby increase Vc use.11,18,20 Surprisingly, there were no TET2 mutations identified in our cohort of patients with AML, but 4 patients had mutations in DNMT3A. Because DNMT3A and TET2 play opposing roles in epigenetic regulation, mutations in DNMT3A alter the activity of TET enzymes, with subsequent changes to Vc use.21 Future studies measuring TET activity may reveal whether epigenetic dysregulation contributes to decreased Vc levels and elucidate possible mechanisms by which Vc supplementation may enhance antitumor immune responses or synergize with hypomethylating agents to improve outcomes in patients with AML.11,22,23 

Contribution: H.C.M., A.C., R.G.M., A.P., and M.R. designed the research; H.C.M. and R.D.S. analyzed and interpreted the data; H.C.M. wrote the manuscript; and all authors collected data and contributed to editing the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Heather C. Milbar, Department of Dermatology, University of Pennsylvania, Perelman Center for Advanced Medicine, 3400 Civic Center Blvd, Philadelphia, PA 19104; e-mail: heather.milbar@pennmedicine.upenn.edu.

1.
Plevin
D
,
Plevin
D
,
Plevin
D
,
Galletly
C
.
The neuropsychiatric effects of vitamin C deficiency: a systematic review
.
BMC Psychiatr
.
2020
;
20
(
1
):
315
.
2.
Velandia
B
,
Centor
RM
,
Mcconnell
V
,
Shah
M
.
.
Scurvy is still present in developed countries
.
2008
;
8
:
1281
-
1284
.
3.
Waldo
AL
,
Zipf
RE
.
Ascorbic acid level in leukemic patients
.
Cancer
.
1955
;
8
(
1
):
187
-
190
.
4.
Huijskens
MJAJ
,
Wodzig
WKWH
,
Walczak
M
,
Germeraad
WTV
,
Bos
GMJ
.
Ascorbic acid serum levels are reduced in patients with hematological malignancies
.
Results Immunol
.
2016
;
6
:
8
-
10
.
5.
Chen
J
,
Nie
D
,
Wang
X
, et al
.
Enriched clonal hematopoiesis in seniors with dietary vitamin C inadequacy
.
Clin Nutr ESPEN
.
2021
;
46
:
179
-
184
.
6.
Loria
CM
,
Klag
MJ
,
Caulfield
LE
,
Whelton
PK
.
Vitamin C status and mortality in US adults
.
Am J Clin Nutr
.
2000
;
72
(
1
):
139
-
145
.
7.
Neyestani
TR
,
Fereydouni
Z
,
Hejazi
S
, et al
.
Vitamin C status in Iranian children with acute lymphoblastic leukemia: evidence for increased utilization
.
J Pediatr Gastroenterol Nutr
.
2007
;
45
(
1
):
141
-
144
.
8.
Vera
JC
,
Rivas
CI
,
Zhang
RH
,
Farber
CM
,
Golde
DW
.
Human HL-60 myeloid leukemia cells transport dehydroascorbic acid via the glucose transporters and accumulate reduced ascorbic acid
.
Blood
.
1994
;
84
(
5
):
1628
-
1634
.
9.
Foster
MN
,
Carr
AC
,
Antony
A
,
Peng
S
,
Fitzpatrick
MG
.
Intravenous vitamin C administration improved blood cell counts and health-related quality of life of patient with history of relapsed acute myeloid leukaemia
.
Antioxidants
.
2018
;
7
(
7
):
92
.
10.
Park
CH
,
Kimler
BF
,
Yi
SY
, et al
.
Depletion of L-ascorbic acid alternating with its supplementation in the treatment of patients with acute myeloid leukemia or myelodysplastic syndromes
.
Eur J Haematol
.
2009
;
83
(
2
):
108
-
118
.
11.
Yue
X
,
Rao
A
.
TET family dioxygenases and the TET activator vitamin C in immune responses and cancer
.
Blood
.
2020
;
136
(
12
):
1394
-
1401
.
12.
Gardner
A
,
Mattiuzzi
G
,
Faderl
S
, et al
.
Randomized comparison of cooked and noncooked diets in patients undergoing remission induction therapy for acute myeloid leukemia
.
J Clin Oncol
.
2008
;
26
(
35
):
5684
-
5688
.
13.
Udensi
UK
,
Tchounwou
PB
.
Dual effect of oxidative stress on leukemia cancer induction and treatment
.
J Exp Clin Cancer Res
.
2014
;
33
:
106
.
14.
Park
S
,
Han
SS
,
Park
CH
, et al
.
L-Ascorbic acid induces apoptosis in acute myeloid leukemia cells via hydrogen peroxide-mediated mechanisms
.
Int J Biochem Cell Biol
.
2004
;
36
(
11
):
2180
-
2195
.
15.
Park
S
,
Park
CH
,
Hahm
ER
, et al
.
Activation of Raf1 and the ERK pathway in response to l-ascorbic acid in acute myeloid leukemia cells
.
Cell Signal
.
2005
;
17
(
1
):
111
-
119
.
16.
Carr
AC
,
Vissers
MCM
,
Cook
JS
.
The effect of intravenous vitamin C on cancer- and chemotherapy-related fatigue and quality of life
.
Front Oncol
.
2014
;
4
:
283
.
17.
Cimmino
L
,
Dolgalev
I
,
Wang
Y
, et al
.
Restoration of TET2 function blocks aberrant self-renewal and leukemia progression
.
Cell
.
2017
;
170
(
6
):
1079
-
1095.e20
.
18.
Agathocleous
M
,
Meacham
CE
,
Burgess
RJ
, et al
.
Ascorbate regulates haematopoietic stem cell function and leukaemogenesis
.
Nature
.
2017
;
549
(
7673
):
476
-
481
.
19.
Shih
AH
,
Abdel-Wahab
O
,
Patel
JP
,
Levine
RL
.
The role of mutations in epigenetic regulators in myeloid malignancies
.
Nat Rev Cancer
.
2012
;
12
(
9
):
599
-
612
.
20.
Guan
Y
,
Greenberg
EF
,
Hasipek
M
, et al
.
Modulation of TET2 activity by ascorbic acid and factors affecting lysine acetylation
.
Blood
.
2018
;
132
(
suppl 1
):
4346
.
21.
López-Moyado
IF
,
Rao
A
.
DNMT3A and TET2 mutations reshape hematopoiesis in opposing ways
.
Nat Genet
.
2020
;
52
(
6
):
554
-
556
.
22.
Zhao
H
,
Zhu
H
,
Huang
J
, et al
.
The synergy of Vitamin C with decitabine activates TET2 in leukemic cells and significantly improves overall survival in elderly patients with acute myeloid leukemia
.
Leuk Res
.
2018
;
66
:
1
-
7
.
23.
Gillberg
L
,
Ørskov
AD
,
Nasif
A
, et al
.
Oral vitamin C supplementation to patients with myeloid cancer on azacitidine treatment: normalization of plasma vitamin C induces epigenetic changes
.
Clin Epigenetics
.
2019
;
11
(
1
):
143
.

Author notes

Data are available on request from the corresponding author, Heather C. Milbar (heather.milbar@pennmedicine.upenn.edu).