Vitamin D deficiency impairs prognosis in many types of cancer; however, its significance in each subtype of hematological malignancies is unclear. In addition, data on the association between pretransplant vitamin D levels and outcomes of hematopoietic stem cell transplantation (HSCT) are inconsistent. This systematic review and meta-analysis aimed to elucidate the impact of vitamin D levels at diagnosis or pre-HSCT on the prognosis of hematological malignancies. Thirty articles and abstracts were extracted from PubMed, Embase, and Cochrane Library databases and conference proceedings. Fixed and random effect models were used to analyze primary outcomes: overall survival (OS) and progression-free survival (PFS). Lower vitamin D level was significantly associated with poorer OS and PFS in myeloid malignancies (hazard ratio [HR], 1.39; 95% confidence interval [CI], 1.06-1.82 and HR, 2.03; 95% CI, 1.23-3.32, respectively) and lymphoid malignancies (HR, 2.07; 95% CI, 1.79-2.40 and HR, 1.91; 95% CI, 1.61-2.25, respectively), as well as outcomes for several lymphoma subtypes individually. Furthermore, a lower pretransplant vitamin D level was associated with poorer OS in autologous and allogeneic HSCT (HR, 1.65; 95% CI, 1.04-2.61 and HR, 1.50; 95% CI, 1.03-2.18, respectively). Despite the relatively small number of studies evaluated, these data suggest the importance of vitamin D status in outcomes of hematological malignancies (PROSPERO registration number: CRD42020205821).

Vitamin D is produced in sun-exposed skin or taken in from the diet, hydroxylated in the liver and the proximal renal tubule to 1,25(OH)2D3, and acts as a steroid hormone by binding to the vitamin D receptor.1  It plays an important role in skeletal health, as well as in tumorigenesis by controlling cell proliferation, apoptosis, differentiation, angiogenesis, invasive and metastatic potential, and tumor immunity.2-4  The association between circulating vitamin D levels and cancer outcomes has been investigated in many types of cancer, and some meta-analyses revealed that higher vitamin D levels result in better outcomes in several cancers, including colorectal,5  breast,5  and prostate cancer6  and melanoma.7 

The role of vitamin D in hematological malignancies has also been studied in clinical settings, because in vitro analysis showed the ability of vitamin D to induce differentiation of human acute myeloid leukemia (AML) cells into mature myeloid cells.8  Although clinically meaningful data using vitamin D and its analogs as differentiation therapy for AML are limited,9  some meta-analyses have revealed that vitamin D deficiency in hematological malignancies was associated with poorer prognosis.5,10  Hematological malignancies include many subtypes of myeloid and lymphoid malignancies; thus, the influence of vitamin D on each subtype should be examined separately, but a detailed analysis to address this issue has not been performed.

In addition, the effects of vitamin D levels on autologous and allogeneic hematopoietic stem cell transplantation (HSCT) have been assessed in several studies, and the significance remains controversial11 ; thus, a comprehensive analysis is warranted. We performed a systematic review and meta-analysis to determine the impact of vitamin D level at diagnosis or pre-HSCT on the prognosis of each subtype of hematological malignancies. This is the first meta-analysis focusing on each subtype of lymphoid malignancies, as well as examining transplant outcomes.

Search strategy

This study was registered with PROSPERO (CRD42020205821) and conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines.12  PubMed, EMBASE, and the COCHRANE registry of clinical trials databases were searched through 17 February 2021 without language restriction, using the following terms: “vitamin D” AND (“lymphoma” OR “leukemia” OR “myeloma” OR “myelodysplastic syndrome” OR “hematological malignancy” OR “hematopoietic stem cell transplantation” OR “bone marrow transplantation”) AND (“progression-free survival” OR “overall survival” OR “PFS” OR “OS” OR “survival” OR “prognosis”). We also searched conference proceedings of the American Society of Hematology (2004-2020), the American Society of Clinical Oncology (2011-2020), and the European Hematology Association (2009-2020) and scanned references of identified articles and reviews for further studies.10,11 

Study selection and quality assessment

Two authors (Y.I. and A.H.) independently assessed the titles and abstracts of all of the identified studies by searching electronic databases. Subsequently, we screened the full texts of the potentially eligible articles. We excluded studies that lacked information needed to evaluate hazard ratios (HRs) of outcomes, were duplicate publications using overlapping patient cohorts, or included many nonmalignant patients undergoing allogeneic HSCT (allo-HSCT). Any discrepancies between the authors were resolved through a discussion including a third author (M.K.) until consensus was reached. The outline of the data extraction is described in Figure 1. The Newcastle-Ottawa scale was used to assess the quality of the nonrandomized trials.13 

Figure 1.

Preferred Reporting Items For Systematic Reviews And Meta-Analysis (PRISMA) flow diagram of study selection. After screening the titles and abstracts of 1212 articles, 52 articles were considered relevant. Among them, 22 articles were excluded for various reasons; 30 articles were included in the analysis. ASCO, American Society of Clinical Oncology; ASH, American Society of Hematology, EHA, European Hematology Society.

Figure 1.

Preferred Reporting Items For Systematic Reviews And Meta-Analysis (PRISMA) flow diagram of study selection. After screening the titles and abstracts of 1212 articles, 52 articles were considered relevant. Among them, 22 articles were excluded for various reasons; 30 articles were included in the analysis. ASCO, American Society of Clinical Oncology; ASH, American Society of Hematology, EHA, European Hematology Society.

Close modal

End points

The primary outcomes in this review were HRs of overall survival (OS) and progression-free survival (PFS), alternatively termed event-free survival, relapse-free survival, or leukemia-free survival in some articles. Secondary outcomes were time-to-treatment (TTT) for patients with chronic lymphocytic leukemia (CLL), relapse rate, and nonrelapse mortality (NRM) for allo-HSCT. When an article reported univariate and multivariate analyses, multivariate data were preferred. When HR was not available, it was estimated using the methods described by Tierney et al.14  When the cohort was divided into >2 groups according to vitamin D level, the data comparing groups with the highest and lowest levels were used. With regard to the measurement of vitamin D level, serum 25-hydroxyvitamin D [25(OH)D] is the major circulating vitamin D metabolite and is used to assess vitamin D status in this meta-analysis; 1 ng/mL of vitamin D corresponds to 2.5 nmol/L.

Statistical analysis

Statistical analyses were performed using EZR software.15  For each trial, the impact of vitamin D deficiency was calculated using HRs with 95% confidence intervals (CIs); mean HRs and the upper 95% CI from each study were input into EZR software for statistical analysis. An HR > 1 favored the higher vitamin D arm. We used the random effect model according to the method of DerSimonian and Laird.16  When the P value for heterogeneity exceeded .10, we preferred the Mantel-Haenszel (fixed effect) method. We assessed the trial results using the χ2 test of heterogeneity and the I2 measure of inconsistency. Heterogeneity was considered statistically significant at P < .10 or I2 > 50%. Potential sources of heterogeneity were investigated using subgroup analyses. Publication bias was examined using funnel plots, coupled with Egger’s test.

Study selection

A literature search of PubMed, EMBASE, and Cochrane Library databases and 3 conference proceedings (American Society of Hematology, American Society of Clinical Oncology, and European Hematology Association) identified 1212 articles after duplicates were eliminated, of which 52 were considered relevant through the evaluation of titles and abstracts. Among them, 30 articles fulfilled the criteria for this meta-analysis: 5 articles on myeloid malignancies,17-21  20 articles on lymphoid malignancies,18,22-40  3 articles on autologous HSCT (ASCT),41-43  and 3 articles on allo-HSCT.44-46  A flow diagram of the article selection process is shown in Figure 1. Twenty-two articles were excluded for the following reasons: duplicate publications from overlapping cohorts,47,48  insufficient data about primary end points,49-60  and transplantations including many nonmalignancies or unidentified diseases.61-68  The characteristics of each study are summarized in Table 1.

Table 1.

Characteristics of the studies included in the meta-analysis

StudyStudy periodCountryDiseaseAge, median (range), yTotal NLow NHigh NMid NVitamin D thresholdMeasuring methodMedian f/uOutcomeNOS
Hu et al, 202017  2014-2016 China AML in the elderly (w/o APL, secondary AML) 70 (60-89) 68 25 43 8.84 ng/mL ECL 2 y OS 
Jackmann et al, 202018  1990-2016 Sweden ALL in children 7 (0.4-17.8) 232 71 161 50 nmol/L CLIA n.a. OS 
   AML in children  52 22 30      
   CML/JMML in children  11 
Radujkovic et al, 201719  2006-2014 Germany MDS, oligoblastic AML 69 (31-83) 58 29 29 32.6 nmol/L CLIA 29 mo OS 
Lee et al, 201420  n.a. US AML (w/o APL) 60 (19-91) 97 29 34 34 <20 ng/mL, ≥32 ng/mL RIA 15.6 mo OS, RFS 
Pardanani et al, 201121  n.a. US PMF 63 (14-83) 247 118 129 25 ng/mL LC-MS/MS 34 mo OS, LFS 
   MDS 72 (44-89) 74 21 53 25 mo 
Mao et al, 202122  2014-2019 China ENKTL 55 (21-92) 93 55 38 50 nmol/L ECL 23 mo OS, PFS 
Wang et al, 202023  2016-2018 China DLBCL 58 (19-85) 208 142 66 52.5 nmol/L ECL 29 mo OS, PFS 
Chen et al, 202133  2011-2018 China DLBCL n.a. 332 111 111 110 <11.5 ng/mL, ≥18.7 ng/mL LC-MS/MS 34.2 mo OS, PFS 
Xu et al, 202034  2014-2018 China MCL 61 (39-77) 70 40 30 50 nmol/L ECL 25.5 mo OS, PFS 
Yellapragada et al, 202035  n.a. US MM 68.9 1889 582 1307 20 ng/mL n.a. n.a. OS 
Borchmann et al, 201936  1993-1998 Germany HL 32 (16-75) 351 175 176 30 nmol/L ELISA 13 y OS, PFS 
Kim et al, 201837  2008-2016 Korea PTCL, ENKTL 17-85 251 105 146 10 ng/mL LC-MS/MS 35.8 mo PFS 
Djurasinovic et al, 201838  2014- 2016 Serbia Lymphoid malignancy (DLBCL, FL, CLL/SLL, HL) 58 (18-84) 133 n.a. n.a. 19.6 nmol/L CLIA 20 mo PFS 
Hohaus et al, 201839  2013-2016 Italy Aggressive BCL 65 154 104 50 20 ng/mL CLIA n.a. EFS 
Ferrari et al, 201740  2013-2016 Italy DLBCL 70 (24-93) 50 n.a. n.a. 15 ng/mL n.a. 20 mo OS, PFS 
Tracy et al, 201724  2002-2012 US FL 60 (23-93) 642 120 522 20 ng/mL LC-MS/MS 59 mo OS 
Cuccaro et al, 201725  2014-2016 Italy HL 33 76 67 10 ng/mL CLIA 12 mo PFS 
Kelly et al, 201526  1998-2008 US FL (SWOG cohort) n.a. 183 28 155 20 ng/mL LC-MS/MS 5.4 y PFS, OS 
 2004-2007 — FL (LYSA cohort) n.a. 240 60 180 10 ng/mL 6.6 y 
Bittenbring et al, 201427  2000-2005 Germany DLBCL with R #1 in the elderly 61-80 184 81 103 8 ng/mL CLIA 34.5 mo PFS, OS 
   DLBCL without R in the elderly 61-80 175 70 105 
 2005-2007  DLBCL with R #2 in the elderly 61-80 63 54   39 mo   
Aref et al, 201328  n.a. Egypt B-CLL 57 (50-60) 75 54 21 20 ng/mL ELISA n.a. OS 
   NHL (DLBCL, MCL, FL, LPL, BL) 61 (52-67) 120 64 56   5 y   
Molica et al, 201229  1998-2008 Italy CLL 68 (43-87) 130 n.a. n.a. 13.5 ng/mL CLIA 39 m TTT 
Tretli et al, 201230  1984-2008 Norway Lymphoma 56.3 (37-79) 145 40 28 77 <46 nmol/L, >81 nmol/L RIA n.a. OS 
Shanafelt et al, 201131  1994-2008 US CLL n.a. 543 180 363 25 ng/mL LC-MS/MS n.a. OS, TTT 
Drake et al, 201032  2002-2008 US NHL (DLBCL, TCL, MCL, FL, other) 62 (19-94) 983 436 547 25 ng/mL LC-MS/MS 34.8 mo EFS, OS 
Eicher et al, 202041  2012-2018 Switzerland ASCT (lymphoma, myeloma) 60 (24-77) 183 102 81 52 nmol/L CLIA n.a. OS 
Rakhee et al, 201642  2010-2015 US ASCT (myeloma) n.a. 158 94 64 23 ng/mL n.a. n.a. OS 
Clairmont et al, 201443  2009- 2010 US ASCT (lymphoma, myeloma) n.a. 132 n.a. n.a. n.a. n.a. n.a. OS 
Bajwa et al, 201944  2012-2017 US Allo-HSCT in children n.a. n.a. 48 78 n.a. ≤20 ng/mL, >30 ng/mL n.a. n.a. OS 
Radujkovic et al, 201745  2002-2013 Germany Allo-HSCT (myeloid #1) 17-75 242 188 54 20 ng/mL CLIA 51.2 mo OS, RR, NRM 
   Allo-HSCT (lymphoid) 17-75 250 208 42      
 2009-2013  Allo-HSCT (myeloid #2) 17-73 398 348 50 51.3 mo 
von Bahr et al, 201546  2005-2011 Sweden Allo-HSCT 12-68 166 19 59 88 <25 nmol/L, ≥50 nmol/L CLIA 71 mo OS 
StudyStudy periodCountryDiseaseAge, median (range), yTotal NLow NHigh NMid NVitamin D thresholdMeasuring methodMedian f/uOutcomeNOS
Hu et al, 202017  2014-2016 China AML in the elderly (w/o APL, secondary AML) 70 (60-89) 68 25 43 8.84 ng/mL ECL 2 y OS 
Jackmann et al, 202018  1990-2016 Sweden ALL in children 7 (0.4-17.8) 232 71 161 50 nmol/L CLIA n.a. OS 
   AML in children  52 22 30      
   CML/JMML in children  11 
Radujkovic et al, 201719  2006-2014 Germany MDS, oligoblastic AML 69 (31-83) 58 29 29 32.6 nmol/L CLIA 29 mo OS 
Lee et al, 201420  n.a. US AML (w/o APL) 60 (19-91) 97 29 34 34 <20 ng/mL, ≥32 ng/mL RIA 15.6 mo OS, RFS 
Pardanani et al, 201121  n.a. US PMF 63 (14-83) 247 118 129 25 ng/mL LC-MS/MS 34 mo OS, LFS 
   MDS 72 (44-89) 74 21 53 25 mo 
Mao et al, 202122  2014-2019 China ENKTL 55 (21-92) 93 55 38 50 nmol/L ECL 23 mo OS, PFS 
Wang et al, 202023  2016-2018 China DLBCL 58 (19-85) 208 142 66 52.5 nmol/L ECL 29 mo OS, PFS 
Chen et al, 202133  2011-2018 China DLBCL n.a. 332 111 111 110 <11.5 ng/mL, ≥18.7 ng/mL LC-MS/MS 34.2 mo OS, PFS 
Xu et al, 202034  2014-2018 China MCL 61 (39-77) 70 40 30 50 nmol/L ECL 25.5 mo OS, PFS 
Yellapragada et al, 202035  n.a. US MM 68.9 1889 582 1307 20 ng/mL n.a. n.a. OS 
Borchmann et al, 201936  1993-1998 Germany HL 32 (16-75) 351 175 176 30 nmol/L ELISA 13 y OS, PFS 
Kim et al, 201837  2008-2016 Korea PTCL, ENKTL 17-85 251 105 146 10 ng/mL LC-MS/MS 35.8 mo PFS 
Djurasinovic et al, 201838  2014- 2016 Serbia Lymphoid malignancy (DLBCL, FL, CLL/SLL, HL) 58 (18-84) 133 n.a. n.a. 19.6 nmol/L CLIA 20 mo PFS 
Hohaus et al, 201839  2013-2016 Italy Aggressive BCL 65 154 104 50 20 ng/mL CLIA n.a. EFS 
Ferrari et al, 201740  2013-2016 Italy DLBCL 70 (24-93) 50 n.a. n.a. 15 ng/mL n.a. 20 mo OS, PFS 
Tracy et al, 201724  2002-2012 US FL 60 (23-93) 642 120 522 20 ng/mL LC-MS/MS 59 mo OS 
Cuccaro et al, 201725  2014-2016 Italy HL 33 76 67 10 ng/mL CLIA 12 mo PFS 
Kelly et al, 201526  1998-2008 US FL (SWOG cohort) n.a. 183 28 155 20 ng/mL LC-MS/MS 5.4 y PFS, OS 
 2004-2007 — FL (LYSA cohort) n.a. 240 60 180 10 ng/mL 6.6 y 
Bittenbring et al, 201427  2000-2005 Germany DLBCL with R #1 in the elderly 61-80 184 81 103 8 ng/mL CLIA 34.5 mo PFS, OS 
   DLBCL without R in the elderly 61-80 175 70 105 
 2005-2007  DLBCL with R #2 in the elderly 61-80 63 54   39 mo   
Aref et al, 201328  n.a. Egypt B-CLL 57 (50-60) 75 54 21 20 ng/mL ELISA n.a. OS 
   NHL (DLBCL, MCL, FL, LPL, BL) 61 (52-67) 120 64 56   5 y   
Molica et al, 201229  1998-2008 Italy CLL 68 (43-87) 130 n.a. n.a. 13.5 ng/mL CLIA 39 m TTT 
Tretli et al, 201230  1984-2008 Norway Lymphoma 56.3 (37-79) 145 40 28 77 <46 nmol/L, >81 nmol/L RIA n.a. OS 
Shanafelt et al, 201131  1994-2008 US CLL n.a. 543 180 363 25 ng/mL LC-MS/MS n.a. OS, TTT 
Drake et al, 201032  2002-2008 US NHL (DLBCL, TCL, MCL, FL, other) 62 (19-94) 983 436 547 25 ng/mL LC-MS/MS 34.8 mo EFS, OS 
Eicher et al, 202041  2012-2018 Switzerland ASCT (lymphoma, myeloma) 60 (24-77) 183 102 81 52 nmol/L CLIA n.a. OS 
Rakhee et al, 201642  2010-2015 US ASCT (myeloma) n.a. 158 94 64 23 ng/mL n.a. n.a. OS 
Clairmont et al, 201443  2009- 2010 US ASCT (lymphoma, myeloma) n.a. 132 n.a. n.a. n.a. n.a. n.a. OS 
Bajwa et al, 201944  2012-2017 US Allo-HSCT in children n.a. n.a. 48 78 n.a. ≤20 ng/mL, >30 ng/mL n.a. n.a. OS 
Radujkovic et al, 201745  2002-2013 Germany Allo-HSCT (myeloid #1) 17-75 242 188 54 20 ng/mL CLIA 51.2 mo OS, RR, NRM 
   Allo-HSCT (lymphoid) 17-75 250 208 42      
 2009-2013  Allo-HSCT (myeloid #2) 17-73 398 348 50 51.3 mo 
von Bahr et al, 201546  2005-2011 Sweden Allo-HSCT 12-68 166 19 59 88 <25 nmol/L, ≥50 nmol/L CLIA 71 mo OS 

ALL, acute lymphoblastic leukemia; APL, acute promyelocytic leukemia; BCL, B-cell lymphoma; BL, Burkitt lymphoma; CLIA, chemiluminescence immunoassay; CLL, chronic lymphocytic leukemia; CML, chronic myeloid leukemia; DLBCL, diffuse large B-cell lymphoma; ECL, electrochemiluminescence; EFS, event-free survival; ELISA, enzyme-linked immunosorbent assay; ENKTL, extranodal natural killer/T-cell lymphoma; FL, follicular lymphoma; f/u, follow-up; High N, patient number with high vitamin D level; HL, Hodgkin lymphoma; JMML, juvenile myelomonocytic leukemia; LC-MS/MS, liquid chromatography–tandem mass spectrometry; LFS, leukemia-free survival; Low N, patient number with low vitamin D level; LPL, lymphoplasmacytic lymphoma; MCL, mantle cell lymphoma; MDS, myelodysplastic syndrome; Mid N, patient number with middle vitamin D level; MM, multiple myeloma; n.a., not available; NHL, non-Hodgkin lymphoma; NOS, Newcastle-Ottawa scale; PMF, primary myelofibrosis; PTCL, peripheral T-cell lymphoma; R, rituximab; RFS, relapse-free survival; RIA, radioimmunoassay; RR, relapse rate; SLL, small lymphocytic lymphoma; TCL, T-cell lymphoma; Total N, total patient number; US, United States; w/o, without.

Vitamin D level in myeloid malignancies at diagnosis

The OS data for myeloid malignancies were available in 5 articles17-21  that included 573 patients with AML, chronic myeloid leukemia, juvenile myelomonocytic leukemia, myelodysplastic syndrome, and primary myelofibrosis. Patients with lower vitamin D levels had significantly poorer OS (HR, 1.39; 95% CI, 1.06-1.82) with substantial heterogeneity (I2 = 57%; P = .03; Figure 2A-B). PFS was analyzed in 384 patients from 3 cohorts;20,21  it was significantly poorer in the group with lower vitamin D status (HR, 2.03; 95% CI, 1.23-3.32) without heterogeneity (I2 = 0%, P = .57; Figure 2C).

Figure 2.

Outcomes in myeloid malignancies. HR of OS survival in myeloid malignancies (A) and funnel plot (B). (C) HR of progression-free survival in myeloid malignancies. CML, chronic myeloid leukemia; JMML, juvenile myelomonocytic leukemia; MDS, myelodysplastic syndrome; PMF, primary myelofibrosis; seTE, standard error of treatment estimate; TE, estimated treatment effect.

Figure 2.

Outcomes in myeloid malignancies. HR of OS survival in myeloid malignancies (A) and funnel plot (B). (C) HR of progression-free survival in myeloid malignancies. CML, chronic myeloid leukemia; JMML, juvenile myelomonocytic leukemia; MDS, myelodysplastic syndrome; PMF, primary myelofibrosis; seTE, standard error of treatment estimate; TE, estimated treatment effect.

Close modal

Vitamin D level in lymphoid malignancies at diagnosis

Data for 4502 patients from 14 articles were eligible for the analysis of OS in lymphoid malignancies.18,22-24,26-28,30-34,36,40  OS was significantly poorer in the group with lower vitamin D status (HR, 2.07; 95% CI, 1.79-2.40) without heterogeneity (I2 = 10%; P = .33; Figure 3A). The funnel plot suggested a publication bias (P = .002; Figure 3B). PFS analyzed in 3436 patients from 13 articles22,23,25-27,32-34,36-40  was also significantly poorer in those with lower vitamin D levels (HR, 1.91; 95% CI, 1.61-2.25) with heterogeneity (I2 = 39%; P = .04; Figure 3C). The funnel plot did not show a publication bias (P = .69; Figure 3D).

Figure 3.

Outcomes in lymphoid malignancies. HR of OS in lymphoid malignancies (A) and funnel plot (B). HR of progression-free survival in lymphoid malignancies (C) and funnel plot (D). ALL, acute lymphoblastic leukemia; BCL, B-cell lymphoma; DLBCL, diffuse large B-cell lymphoma; ENKTL, extranodal natural killer/T-cell lymphoma; FL, follicular lymphoma; HL, Hodgkin lymphoma; MCL, mantle cell lymphoma; NHL, non-Hodgkin lymphoma; R, rituximab; seTE, standard error of treatment estimate; TCL, T-cell lymphoma; TE, estimated treatment effect.

Figure 3.

Outcomes in lymphoid malignancies. HR of OS in lymphoid malignancies (A) and funnel plot (B). HR of progression-free survival in lymphoid malignancies (C) and funnel plot (D). ALL, acute lymphoblastic leukemia; BCL, B-cell lymphoma; DLBCL, diffuse large B-cell lymphoma; ENKTL, extranodal natural killer/T-cell lymphoma; FL, follicular lymphoma; HL, Hodgkin lymphoma; MCL, mantle cell lymphoma; NHL, non-Hodgkin lymphoma; R, rituximab; seTE, standard error of treatment estimate; TCL, T-cell lymphoma; TE, estimated treatment effect.

Close modal

Vitamin D level in DLBCL

Next, we analyzed each subtype of lymphoid malignancy. Data on 1272 patients with diffuse large B-cell lymphoma (DLBCL) from 5 articles were eligible for analysis of OS and PFS.23,27,32,33,40  Lower vitamin D status was associated with poorer OS (HR, 2.20; 95% CI, 1.70-2.86) and PFS (HR, 2.05; 95% CI, 1.47-2.86) (Figure 4).

Figure 4.

Outcomes in DLBCL. HR of OS in DLBCL (A) and funnel plot (B). HR of progression-free survival in DLBCL (C) and funnel plot (D). R, rituximab; seTE, standard error of treatment estimate; TE, estimated treatment effect.

Figure 4.

Outcomes in DLBCL. HR of OS in DLBCL (A) and funnel plot (B). HR of progression-free survival in DLBCL (C) and funnel plot (D). R, rituximab; seTE, standard error of treatment estimate; TE, estimated treatment effect.

Close modal

Vitamin D level in FL at diagnosis

For follicular lymphoma (FL), data on 1065 patients from 3 cohorts were eligible for the analysis of OS.24,26  Tracy et al performed an expanded analysis of their prior cohort; thus, we used their expanded data.24,32  Lower vitamin D status was also associated with significantly poorer OS (HR, 2.55; 95% CI, 1.68-3.88) without heterogeneity (Figure 5A). PFS data were available for 423 patients from 2 cohorts;26  lower vitamin D status was associated with significantly poorer PFS (HR, 1.67; 95% CI, 1.16-2.42) without heterogeneity (Figure 5B).

Figure 5.

Outcomes in each subtype of lymphoid malignancies. HRs of OS (A) and PFS (B) in FL. HRs of OS (C) and PFS (D) in MCL. (E) HR of PFS in HL. HRs of OS (F) and PFS (G) in TCL. HRs of OS (H) and TTT (I) in CLL. seTE, standard error of treatment estimate; TE, estimated treatment effect.

Figure 5.

Outcomes in each subtype of lymphoid malignancies. HRs of OS (A) and PFS (B) in FL. HRs of OS (C) and PFS (D) in MCL. (E) HR of PFS in HL. HRs of OS (F) and PFS (G) in TCL. HRs of OS (H) and TTT (I) in CLL. seTE, standard error of treatment estimate; TE, estimated treatment effect.

Close modal

Vitamin D level in MCL at diagnosis

For mantle cell lymphoma (MCL), the data on 141 patients from 2 articles were available32,34 ; they did not show any significant association between vitamin D level and OS (HR, 3.10; 95% CI, 0.53-8.24) or PFS (HR, 1.98; 95% CI, 0.60-6.59) with substantial heterogeneity (Figure 5C-D).

Vitamin D level in HL at diagnosis

For Hodgkin lymphoma (HL), OS data were available for 351 patients from only 1 article,36  which reported that lower vitamin D status was related to impaired OS (HR, 1.82; 95% CI, 1.53-2.15). The data on 427 patients from 2 articles were eligible for analysis of PFS,25,36  which revealed poor PFS in patients with lower vitamin D level (HR, 2.31; 95% CI, 1.36-3.93; Figure 5E).

Vitamin D level in TCL at diagnosis

For T-cell lymphoma (TCL), data for 163 patients from 2 articles22,32  and 414 patients from 3 articles22,32,37  were eligible for the analysis of OS and PFS, respectively. Lower vitamin D status was associated with poor OS and PFS (HR, 2.49; 95% CI, 1.38-4.48 and HR, 1.97; 95% CI, 1.38-2.82, respectively; Figure 5F-G). Kim et al further divided TCL into peripheral TCL and extranodal natural killer/T-cell lymphoma37  and showed that vitamin D deficiency impaired prognosis only in the latter group of patients; thus, further subdivision of TCL may be required for more accurate examination.

Vitamin D level in CLL at diagnosis

For CLL, the data for 618 patients from 2 articles were eligible for the analysis of OS,28,31  which showed that lower vitamin D status was not significantly associated with OS (HR, 2.07; 95% CI, 0.80-5.36; Figure 5H). There were no articles reporting on PFS, and TTT data were available from 2 articles with 673 patients.29,31  TTT was significantly shorter in the group of patients with lower vitamin D status (HR, 1.55; 95% CI, 1.19-2.00; Figure 5I).

Vitamin D level in MM at diagnosis

For multiple myeloma (MM), only 1 article was included in our search;35  it reported that lower vitamin D status was significantly associated with poor OS (HR, 1.34; P = .008). The investigators showed that its impact on OS was observed in white patients (HR, 1.45; P = .005), but not in African Americans, suggesting a difference between ethnic groups.

Threshold of vitamin D level in the studies of lymphoid malignancies

There is no agreed upon cutoff value for the definition of vitamin D deficiency,11  and the studies included in this meta-analysis used various thresholds based on different guidelines (Table 1). To examine this discrepancy, we performed subgroup analyses based on the cutoff values in lymphoid malignancies. Eight studies defined 50 nmol/L (20 ng/mL) as a threshold18,22,24,26,28,34,35,39 ; patients with vitamin D levels less than this value had poorer OS (HR, 2.83; 95% CI, 1.81-4.42) and PFS significantly (HR, 2.48; 95% CI, 1.76-3.51; supplemental Figure 1A-B) without heterogeneity. Three studies defined 25 nmol/L (10 ng/mL) as a threshold;25,26,37  a vitamin D level less than this value was associated with poorer PFS (HR, 1.75; 95% CI, 1.19-2.59; supplemental Figure 1C). Two studies used 62.5 nmol/L (25 ng/mL) as a threshold;31,32  a vitamin D level less than this value was also associated with poorer OS (HR, 1.77; 95% CI, 1.36-2.29) and PFS (HR, 1.32; 95% CI, 1.04-1.68; supplemental Figure 1D-E). All subgroup analyses demonstrated that lower vitamin D levels were associated with poorer outcomes.

The impact of geographical distribution on vitamin D level in lymphoid malignancies

Geographical factors, such as latitude and sunlight exposure, affect the levels of vitamin D in the normal population11 ; thus, we performed subgroup analysis in lymphoid malignancies for the United States,24,26,31,32,35  China,22,23,33,34  Italy,25,29,39,40  and Germany.27,36  A lower vitamin D level was associated with significantly poorer OS (HR, 1.96; 95% CI, 1.56-2.46) and PFS (HR, 1.40; 95% CI, 1.12-1.75) in the United States, significantly poorer OS (HR, 3.26; 95% CI, 2.00-5.30) and PFS (HR, 2.99; 95% CI, 2.12-4.21) in China, significantly poorer PFS (HR, 3.80; 95% CI, 1.90-7.60) in Italy, and significantly poorer OS (HR, 1.85; 95% CI, 1.59-2.15) and PFS (HR, 1.92; 95% CI, 1.54-2.39) in Germany (supplemental Figure 2).

Vitamin D level at ASCT

We then focused on the significance of vitamin D levels during HSCT. First, we analyzed the data from patients with lymphoma and myeloma who underwent ASCT. The data for 141 lymphoma patients and 332 myeloma patients from 3 articles were eligible for analysis of OS;41-43  lower vitamin D status was associated with significantly poorer OS (HR, 1.65; 95% CI, 1.04-2.61, using the fixed effect model), with low heterogeneity (I2 = 21%, P = .28) (Figure 6A).

Figure 6.

Outcomes in HSCT. (A) HR of OS in ASCT. HRs of OS (B), relapse rate (C), and NRM (D) in allo-HSCT. seTE, standard error of treatment estimate; TE, estimated treatment effect.

Figure 6.

Outcomes in HSCT. (A) HR of OS in ASCT. HRs of OS (B), relapse rate (C), and NRM (D) in allo-HSCT. seTE, standard error of treatment estimate; TE, estimated treatment effect.

Close modal

Vitamin D level at allo-HSCT

Next, we extracted allo-HSCT data for hematological malignancies. The data for 1094 patients from 5 cohorts were eligible for the analysis of OS.44 -46  Bajwa et al analyzed OS in patients with malignancy and nonmalignancy separately; thus, we only used data for patients with malignancy.44  The meta-analysis showed that lower vitamin D status was associated with significantly poorer OS (HR, 1.50; 95% CI, 1.03-2.18; Figure 6B). Data for relapse rate and NRM were available from 3 cohorts in only 1 article45  that included 890 patients; lower vitamin D status was related to a high relapse rate (HR, 2.12; 95% CI, 1.41-3.19) but not to NRM (HR, 1.23; 95% CI, 0.72-2.10; Figure 6C-D).

This meta-analysis comprehensively investigated the impact of circulating vitamin D levels at diagnosis on the prognosis of hematological malignancies and each subset of lymphoid malignancy. We showed that lower vitamin D level at diagnosis was related to a significantly impaired prognosis for myeloid and lymphoid malignancies, as previously reported.5,10  Moreover, further subgroup analysis revealed that a lower vitamin D level was associated with a poorer prognosis for several lymphoma subtypes: DLBCL, FL, HL, and TCL. Although the survival data for patients with MCL or CLL did not reach statistical significance, lower vitamin D status shortened TTT in CLL.

Vitamin D is associated with bone homeostasis, as well as with tumorigenesis via many mechanisms.2,3,11  Several reports have confirmed the direct antitumor effect of vitamin D against leukemia and lymphoma cells in vitro: an antiproliferation effect in non-HL69  and myeloma70  and induction of apoptosis in B-cell CLL71 . Vitamin D also exerts synergistic effects with other anticancer agents (eg, azacytidine) against myeloid cell lines19  and etoposide and doxorubicin against HL cell lines.36  Moreover, vitamin D potentiates antitumor immunity by activating natural killer cells27  and macrophages72 ; thus, vitamin D has a variety of protective mechanisms against hematological malignancies, and further investigation is warranted to understand the entire spectrum.73 

Several meta-analyses have been performed to elucidate the role of vitamin D in hematological malignancies (Table 2). Vitamin D deficiency was consistently associated with poorer OS and PFS in leukemia and lymphoma patients,5,10,74  Also, some meta-analyses focused on the relationship between vitamin D status and the risk of lymphoma but did not find any significant correlation.75-77 

Table 2.

Characteristics of previous meta-analyses of the role of vitamin D status in hematological malignancies

StudyDiseaseNo. of articles includedHR (95% CI)
This study Myeloid malignancy OS, 1.39 (1.06-1.82); PFS, 2.03 (1.23-3.32) 
 Lymphoid malignancy 20 OS, 2.07 (1.79-2.40); PFS, 1.91 (1.61-2.25) 
 ASCT OS, 1.65 (1.04-2.61) 
 Allo-HSCT OS, 1.50 (1.03-2.18) 
Tao et al, 202174  Lymphoma 12 OS, 1.94 (1.71-2.19); PFS, 2.06 (1.82-2.32) 
Chiengthong et al, 202079  HSCT aGVHD, 1.07 (0.74-1.53); cGVHD, 1.75 (0.72-4.26) 
Wang et al, 201510  Hematological cancers OS, 1.85 (1.54-2.23); RFS, 1.45 (1.25-1.70) 
  Leukemia OS, 2.17 (1.54-3.05); RFS, 1.74 (1.34-2.27) 
  Lymphoma OS, 1.95 (1.47-2.59); RFS, 1.25 (1.02-1.54) 
Li et al, 20145  Lymphoma OS, 2.08 (1.56-2.78) 
StudyDiseaseNo. of articles includedHR (95% CI)
This study Myeloid malignancy OS, 1.39 (1.06-1.82); PFS, 2.03 (1.23-3.32) 
 Lymphoid malignancy 20 OS, 2.07 (1.79-2.40); PFS, 1.91 (1.61-2.25) 
 ASCT OS, 1.65 (1.04-2.61) 
 Allo-HSCT OS, 1.50 (1.03-2.18) 
Tao et al, 202174  Lymphoma 12 OS, 1.94 (1.71-2.19); PFS, 2.06 (1.82-2.32) 
Chiengthong et al, 202079  HSCT aGVHD, 1.07 (0.74-1.53); cGVHD, 1.75 (0.72-4.26) 
Wang et al, 201510  Hematological cancers OS, 1.85 (1.54-2.23); RFS, 1.45 (1.25-1.70) 
  Leukemia OS, 2.17 (1.54-3.05); RFS, 1.74 (1.34-2.27) 
  Lymphoma OS, 1.95 (1.47-2.59); RFS, 1.25 (1.02-1.54) 
Li et al, 20145  Lymphoma OS, 2.08 (1.56-2.78) 

aGVHD, acute graft-versus-host disease; cGVHD, chronic graft-versus-host disease; RFS, relapse-free survival.

We also performed the first meta-analysis of the association between pretransplant vitamin D levels and prognosis, which showed a significant negative impact of lower vitamin D status on OS in ASCT and allo-HSCT patients, despite the limited number of studies. Lower vitamin D level at allo-HSCT was not associated with NRM, but it was associated with a higher relapse rate, which might have resulted in a worse prognosis. Studies on pediatric transplantation that included a substantial proportion of nonmalignant diseases were excluded from this meta-analysis. Some of them concluded that vitamin D deficiency was associated with worse outcome62,66 ; thus, subgroup analyses that differentiate between malignant and nonmalignant diseases are needed in the future. Pretransplant vitamin D deficiency has also been implicated in the pathogenesis of graft-versus-host disease via immunomodulatory effects,46,78  but a recent meta-analysis did not show a statistically significant association79  (Table 2).

Evidence for an impact of vitamin D levels on cancer prognosis has been accumulating gradually, but it remains uncertain whether vitamin D supplementation can improve cancer prognosis. A recent meta-analysis demonstrated a clinically meaningful benefit for vitamin D supplementation on colorectal cancer survival outcomes.80  With regard to hematological malignancies, some studies suggested that vitamin D supplementation was associated with improved event-free survival in DLBCL patients39  and relapse-free survival after ASCT.81  Larger randomized clinical trials are needed to establish additional evidence.

This meta-analysis has several limitations. First, the threshold for vitamin D level differed between articles. The threshold is determined based on clinical effects in bone health, such as osteoporosis, bone mineral density, and hip fractures, or the inverse relationship between serum parathyroid hormone and vitamin D level.82,83  Several studies calculated different thresholds using their own clinical data respectively; thus, a consensus has not been achieved.82  For example, Endocrine Society clinical practice guidelines define deficiency as <20 ng/mL, insufficiency as 21 to 29 ng/mL, and sufficiency as >30 ng/mL.84  Another report defines severe deficiency as <5 ng/mL, moderate deficiency as 5 to 10 ng/mL, mild deficiency as 10 to 20 ng/mL, and replete as >20 ng/mL.83  Other reviews suggest 10 ng/mL, 12 ng/mL, 20 ng/mL,11  or 30 ng/mL85  as a cutoff. The definition is different among guidelines and reviews, and the studies in our meta-analyses referred to them individually, resulting in the discrepancies shown in Table 1. Vitamin D levels are also influenced by geographic region, diet, environmental factors, and lifestyle,86  and the optimal target remains unclear.11,26  In MM patients, vitamin D deficiency was associated with poor OS in white, but not in African American,35  patients, suggesting the importance of racial differences. To clarify the significance of these discrepancies, we performed subgroup analysis classified by each cutoff value (25, 50, and 62.5 nmol/L) and each country (United States, China, Italy, and Germany) in lymphoid malignancies, which confirmed the significant relationship between lower vitamin D levels and poorer outcomes in all subgroups. Second, the measurement method was also different between articles. Liquid chromatography-tandem mass spectrometry is recommended as the most accurate method, but immunoassays were used in some articles;87  thus, the measurement method should be agreed upon. Third, the number of studies specific to each subtype of leukemia and lymphoma remains insufficient. With regard to myeloid malignancies, we could not analyze patients with AML, MDS, chronic myeloid leukemia/juvenile myelomonocytic leukemia, or primary myelofibrosis individually. These subtypes are different with regard to disease biology; thus, they should be analyzed separately. Considering lymphoid malignancies, the data on DLBCL patients were relatively abundant: 5 articles, including 7 cohorts, but other lymphoma subgroups were studied in only 1 to 3 articles. In addition, the condition of transplantation is quite different according to the type of disease, disease status, age, donor source, preconditioning regimen, and so on; thus, it should be adjusted in future investigations. Fourth, 25(OH)D is commonly used as a reliable indicator of vitamin D status; however, some articles recommend bioavailable 25(OH)D level as a precise biomarker.33  Moreover, Peter et al showed that peritransplant 1,25(OH)2D3 levels, but not 25(OH)D levels, predicted survival after stem cell transplantation.49  Therefore, the most precise biomarker of vitamin D levels in predicting the outcome of malignancies should be investigated further. Fifth, although our meta-analysis clarified the association between low vitamin D level and poorer PFS and OS, it does not establish causality and cannot exclude the possibility of residual confounding or reverse causality. Sixth, some patients with vitamin D deficiency were prescribed vitamin D supplementation,39  but most articles did not mention whether patients received supplementation. Thus, we could not include this factor in this meta-analysis. However, even if patients with vitamin D deficiency received supplementation, our results still suggest that lower vitamin D status at diagnosis has a worse prognosis. The significance of vitamin D supplementation needs to be addressed in future prospective trials.

In conclusion, this meta-analysis demonstrated that lower vitamin D status at diagnosis was associated with a significantly worse prognosis for myeloid and lymphoid malignancies, as well as several lymphoma subtypes, including DLBCL, FL, HL, and TCL. We also showed that pretransplant vitamin D level was an important factor for prognosis in ASCT and allo-HSCT. Further studies focusing on each subtype of hematological malignancy are warranted.

Contribution: Y.I. conceptualized and designed the study, performed literature research, analyzed data, performed statistical analyses, and wrote the manuscript; A.H. performed literature review and critically revised the manuscript; and M.K. supervised the study.

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

Correspondence: Mineo Kurokawa, Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; e-mail: kurokawa-tky@umin.ac.jp.

1.
Christakos
S
,
Dhawan
P
,
Verstuyf
A
,
Verlinden
L
,
Carmeliet
G
.
Vitamin D metabolism, molecular mechanism of action, and pleiotropic effects
.
Physiol Rev.
2016
;
96
(
1
):
365
-
408
.
2.
Bandera Merchan
B
,
Morcillo
S
,
Martin-Nuñez
G
,
Tinahones
FJ
,
Macías-González
M
.
The role of vitamin D and VDR in carcinogenesis: through epidemiology and basic sciences
.
J Steroid Biochem Mol Biol.
2017
;
167
:
203
-
218
.
3.
Holick
MF
.
Vitamin D deficiency
.
N Engl J Med.
2007
;
357
(
3
):
266
-
281
.
4.
Hall
AC
,
Juckett
MB
.
The role of vitamin D in hematologic disease and stem cell transplantation
.
Nutrients.
2013
;
5
(
6
):
2206
-
2221
.
5.
Li
M
,
Chen
P
,
Li
J
,
Chu
R
,
Xie
D
,
Wang
H
.
Review: the impacts of circulating 25-hydroxyvitamin D levels on cancer patient outcomes: a systematic review and meta-analysis
.
J Clin Endocrinol Metab.
2014
;
99
(
7
):
2327
-
2336
.
6.
Song
ZY
,
Yao
Q
,
Zhuo
Z
,
Ma
Z
,
Chen
G
.
Circulating vitamin D level and mortality in prostate cancer patients: a dose-response meta-analysis
.
Endocr Connect.
2018
;
7
(
12
):
R294
-
R303
.
7.
Tsai
TY
,
Kuo
CY
,
Huang
YC
.
The association between serum vitamin D level and risk and prognosis of melanoma: a systematic review and meta-analysis
.
J Eur Acad Dermatol Venereol.
2020
;
34
(
8
):
1722
-
1729
.
8.
Miyaura
C
,
Abe
E
,
Kuribayashi
T
, et al
.
1 α,25-Dihydroxyvitamin D3 induces differentiation of human myeloid leukemia cells
.
Biochem Biophys Res Commun.
1981
;
102
(
3
):
937
-
943
.
9.
Harrison
JS
,
Bershadskiy
A
.
Clinical experience using vitamin D and analogs in the treatment of myelodysplasia and acute myeloid leukemia: a review of the literature
.
Leukemia Res Treat.
2012
;
2012
:
125814
.
10.
Wang
W
,
Li
G
,
He
X
, et al
.
Serum 25-hydroxyvitamin D levels and prognosis in hematological malignancies: a systematic review and meta-analysis
.
Cell Physiol Biochem.
2015
;
35
(
5
):
1999
-
2005
.
11.
Soto
JR
,
Anthias
C
,
Madrigal
A
,
Snowden
JA
.
Insights into the role of vitamin D as a biomarker in stem cell transplantation
.
Front Immunol.
2020
;
11
:
966
.
12.
Moher
D
,
Liberati
A
,
Tetzlaff
J
,
Altman
DG
;
PRISMA Group
.
Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement
.
Ann Intern Med.
2009
;
151
(
4
):
264
-
269, W64
.
13.
Stang
A
.
Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses
.
Eur J Epidemiol.
2010
;
25
(
9
):
603
-
605
.
14.
Tierney
JF
,
Stewart
LA
,
Ghersi
D
,
Burdett
S
,
Sydes
MR
.
Practical methods for incorporating summary time-to-event data into meta-analysis
.
Trials.
2007
;
8
(
1
):
16
.
15.
Kanda
Y
.
Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics
.
Bone Marrow Transplant.
2013
;
48
(
3
):
452
-
458
.
16.
DerSimonian
R
,
Laird
N
.
Meta-analysis in clinical trials
.
Control Clin Trials.
1986
;
7
(
3
):
177
-
188
.
17.
Hu
B
,
Chen
L
,
Wu
X
, et al
.
A retrospective study of the association of low 25(OH) vitamin D3 levels with poor outcomes in elderly patients with de novo acute myeloid leukaemia
.
Acta Med Mediter.
2020
;
36
(
6
):
3573
-
3578
.
18.
Jackmann
N
,
Mäkitie
O
,
Harila-Saari
A
,
Gustafsson
J
,
Nezirevic Dernroth
D
,
Frisk
P
.
Vitamin D status in children with leukemia, its predictors, and association with outcome
.
Pediatr Blood Cancer.
2020
;
67
(
4
):
e28163
.
19.
Radujkovic
A
,
Schnitzler
P
,
Ho
AD
,
Dreger
P
,
Luft
T
.
Low serum vitamin D levels are associated with shorter survival after first-line azacitidine treatment in patients with myelodysplastic syndrome and secondary oligoblastic acute myeloid leukemia
.
Clin Nutr.
2017
;
36
(
2
):
542
-
551
.
20.
Lee
HJ
,
Muindi
JR
,
Tan
W
, et al
.
Low 25(OH) vitamin D3 levels are associated with adverse outcome in newly diagnosed, intensively treated adult acute myeloid leukemia
.
Cancer.
2014
;
120
(
4
):
521
-
529
.
21.
Pardanani
A
,
Drake
MT
,
Finke
C
, et al
.
Vitamin D insufficiency in myeloproliferative neoplasms and myelodysplastic syndromes: clinical correlates and prognostic studies
.
Am J Hematol.
2011
;
86
(
12
):
1013
-
1016
.
22.
Mao
J
,
Yin
H
,
Wang
L
, et al
.
Prognostic value of 25-hydroxy vitamin D in extranodal NK/T cell lymphoma
.
Ann Hematol.
2021
;
100
(
2
):
445
-
453
.
23.
Wang
WT
,
Liang
JH
,
Wang
L
, et al
.
The prognostic value of 25-hydroxy vitamin D deficiency and its interaction with c-Myc expression in diffuse large B cell lymphoma
.
Ann Hematol.
2020
;
99
(
10
):
2377
-
2384
.
24.
Tracy
SI
,
Maurer
MJ
,
Witzig
TE
, et al
.
Vitamin D insufficiency is associated with an increased risk of early clinical failure in follicular lymphoma
.
Blood Cancer J.
2017
;
7
(
8
):
e595
.
25.
Cuccaro
A
,
Galli
E
,
Visconti
F
, et al
25(OH)Vitamin D serum levels in Hodgkin lymphoma
.
Haematologica.
2017
;
102
:
Supplement 2
(
462
).
26.
Kelly
JL
,
Salles
G
,
Goldman
B
, et al
.
Low serum vitamin D levels are associated with inferior survival in follicular lymphoma: a prospective evaluation in SWOG and LYSA Studies
.
J Clin Oncol.
2015
;
33
(
13
):
1482
-
1490
.
27.
Bittenbring
JT
,
Neumann
F
,
Altmann
B
, et al
.
Vitamin D deficiency impairs rituximab-mediated cellular cytotoxicity and outcome of patients with diffuse large B-cell lymphoma treated with but not without rituximab
.
J Clin Oncol.
2014
;
32
(
29
):
3242
-
3248
.
28.
Aref
S
,
Ibrahim
L
,
Azmy
E
.
Prognostic impact of serum 25-hydroxivitamin D [25(OH)D] concentrations in patients with lymphoid malignancies
.
Hematology.
2013
;
18
(
1
):
20
-
25
.
29.
Molica
S
,
Digiesi
G
,
Antenucci
A
, et al
.
Vitamin D insufficiency predicts time to first treatment (TFT) in early chronic lymphocytic leukemia (CLL)
.
Leuk Res.
2012
;
36
(
4
):
443
-
447
.
30.
Tretli
S
,
Schwartz
GG
,
Torjesen
PA
,
Robsahm
TE
.
Serum levels of 25-hydroxyvitamin D and survival in Norwegian patients with cancer of breast, colon, lung, and lymphoma: a population-based study
.
Cancer Causes Control.
2012
;
23
(
2
):
363
-
370
.
31.
Shanafelt
TD
,
Drake
MT
,
Maurer
MJ
, et al
.
Vitamin D insufficiency and prognosis in chronic lymphocytic leukemia
.
Blood.
2011
;
117
(
5
):
1492
-
1498
.
32.
Drake
MT
,
Maurer
MJ
,
Link
BK
, et al
.
Vitamin D insufficiency and prognosis in non-Hodgkin’s lymphoma
.
J Clin Oncol.
2010
;
28
(
27
):
4191
-
4198
.
33.
Chen
P
,
Cao
Y
,
Duan
X
,
Li
J
,
Zhao
W
,
Wang
H
.
Bioavailable 25(OH)D level is associated with clinical outcomes of patients with diffuse large B-cell lymphoma: an exploratory study
.
Clin Nutr.
2021
;
40
(
1
):
157
-
165
.
34.
Xu
DM
,
Liang
JH
,
Wang
L
, et al
.
25-Hydroxy vitamin D deficiency predicts inferior prognosis in mantle cell lymphoma
.
J Cancer Res Clin Oncol.
2020
;
146
(
4
):
1003
-
1009
.
35.
Yellapragada
SV
,
Fillmore
NR
,
Frolov
A
, et al
.
Vitamin D deficiency predicts for poor overall survival in white but not African American patients with multiple myeloma
.
Blood Adv.
2020
;
4
(
8
):
1643
-
1646
.
36.
Borchmann
S
,
Cirillo
M
,
Goergen
H
, et al
.
Pretreatment Vitamin D deficiency is associated with impaired progression-free and overall survival in Hodgkin lymphoma
.
J Clin Oncol.
2019
;
37
(
36
):
3528
-
3537
.
37.
Kim
SJ
,
Shu
C
,
Ryu
KJ
, et al
.
Vitamin D deficiency is associated with inferior survival of patients with extranodal natural killer/T-cell lymphoma
.
Cancer Sci.
2018
;
109
(
12
):
3971
-
3980
.
38.
Djurasinović
VT
,
Mihaljević
BS
,
Šipetić Grujičić
SB
, et al
.
25(OH) vitamin D deficiency in lymphoid malignancies, its prevalence and significance. Are we fully aware of it?
Support Care Cancer.
2018
;
26
(
8
):
2825
-
2832
.
39.
Hohaus
S
,
Tisi
MC
,
Bellesi
S
, et al
.
Vitamin D deficiency and supplementation in patients with aggressive B-cell lymphomas treated with immunochemotherapy
.
Cancer Med.
2018
;
7
(
1
):
270
-
281
.
40.
Ferrari
A
,
Ruffini
A
,
Alvarez
I
, et al
.
The role of vitamin D as a prognostic factor in diffuse large B cell lymphoma: a monocentric study from hematology unit of Reggio Emilia
.
Haematologica.
2017
;
102
:Supplement 3 (
114
).
41.
Eicher
F
,
Mansouri Taleghani
B
,
Schild
C
,
Bacher
U
,
Pabst
T
.
Reduced survival after autologous stem cell transplantation in myeloma and lymphoma patients with low vitamin D serum levels
.
Hematol Oncol.
2020
;
38
(
4
):
523
-
530
.
42.
Rakhee
V
,
Ahlers
S
,
Rodriguez
C
, et al
.
Low pre-transplant vitamin D levels predict an inferior survival in patients with multiple myeloma undergoing an autologous stem cell transplant
.
Blood.
2016
;
128
(
22
):
5655
.
43.
Clairmont
EB
,
Schoch
G
,
Gopal
AK
,
McDonnell
P
.
25-hydroxyvitamin D concentrations and overall survival in autologous hematopoietic stem cell subjects
.
Biol Blood Marrow Transplant.
2014
;
20
(
2
):
S106
-
S107
.
44.
Bajwa
RP
,
Taylor
K
,
Hoyt
AR
, et al
.
Effects of vitamin D levels on outcomes after allogeneic hematopoietic stem cell transplantation in children
.
Biol Blood Marrow Transplant.
2019
;
25
(
3
):
S239
-
S240
.
45.
Radujkovic
A
,
Kordelas
L
,
Krzykalla
J
, et al
.
Pretransplant vitamin D deficiency is associated with higher relapse rates in patients allografted for myeloid malignancies
.
J Clin Oncol.
2017
;
35
(
27
):
3143
-
3152
.
46.
von Bahr
L
,
Blennow
O
,
Alm
J
, et al
.
Increased incidence of chronic GvHD and CMV disease in patients with vitamin D deficiency before allogeneic stem cell transplantation
.
Bone Marrow Transplant.
2015
;
50
(
9
):
1217
-
1223
.
47.
Djurasinovic
V
,
Mihaljevic
B
,
Vukovic
V
, et al
.
Pretreatment serum 25(OH) vitamin D as predictor of event- free survival in lymphoid malignancies
.
HemaSphere.
2019
;
3
(
S1
):
1018
.
48.
Robsahm
TE
,
Tretli
S
,
Torjesen
PA
,
Babigumira
R
,
Schwartz
GG
.
Serum 25-hydroxyvitamin D levels predict cancer survival: a prospective cohort with measurements prior to and at the time of cancer diagnosis
.
Clin Epidemiol.
2019
;
11
:
695
-
705
.
49.
Peter
K
,
Siska
PJ
,
Roider
T
, et al
.
1,25-dihydroxyvitamin-D3 but not the clinically applied marker 25-hydroxyvitamin-D3 predicts survival after stem cell transplantation
.
Bone Marrow Transplant.
2021
;
56
(
2
):
419
-
433
.
50.
Nair
R
,
Hadidi
SA
,
Steiner
RE
, et al
.
Association of vitamin D deficiency with inferior treatment outcomes in patients with newly diagnosed classic Hodgkin Lymphoma: MD Anderson Cancer Center Experience
.
Blood.
2020
;
136
(
suppl 1
):
27
-
28
.
51.
Thomas
X
,
Chelghoum
Y
,
Fanari
N
,
Cannas
G
.
Serum 25-hydroxyvitamin D levels are associated with prognosis in hematological malignancies
.
Hematology.
2011
;
16
(
5
):
278
-
283
.
52.
Ng
AC
,
Kumar
SK
,
Rajkumar
SV
,
Drake
MT
.
Impact of vitamin D deficiency on the clinical presentation and prognosis of patients with newly diagnosed multiple myeloma
.
Am J Hematol.
2009
;
84
(
7
):
397
-
400
.
53.
Sanchez-Gonzalez
B
,
Platero
J
,
Ortuño
A
, et al
.
Vitamin D insufficiency and prognosis in lymphoma
.
HemaSphere.
2019
;
3
(
S1
):
115
-
116
.
54.
Seyedalipour
F
,
Mansouri
A
,
Vaezi
M
, et al
.
High prevalence of vitamin D deficiency in newly diagnosed acute myeloid leukemia patients and its adverse outcome
.
Int J Hematol Oncol Stem Cell Res.
2017
;
11
(
3
):
209
-
216
.
55.
Lad
DP
,
Mourad
YA
,
Barnett
MJ
, et al
.
Pre-transplant vitamin D deficiency is associated with inferior overall survival but not associated with relapse free survival or cumulative incidence of GVHD post adult hematopoietic cell transplantation for hematological malignancies
.
Biol Blood Marrow Transplant.
2016
;
22
(
3
):
S334
.
56.
Lauter
B
,
Schmidt-Wolf
IGH
.
Prevalence, supplementation, and impact of vitamin D deficiency in multiple myeloma patients
.
Cancer Invest.
2015
;
33
(
10
):
505
-
509
.
57.
Hudzik
S
,
Snoad
B
,
Mousa
L
, et al
.
The majority of myeloma patients are vitamin D deficient, unrelated to survival or cytogenetics
.
Blood.
2015
;
126
(
23
):
5336
.
58.
Ganetsky
A
,
Richman
LP
,
Frey
NV
, et al
.
Vitamin d deficiency predicts acute cutaneous graft-versus-host disease in reduced-intensity allogeneic hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant.
2014
;
20
(
2
):
S267
-
S268
.
59.
Glotzbecker
B
,
Ho
VT
,
Aldridge
J
, et al
.
Low levels of 25-hydroxyvitamin D before allogeneic hematopoietic SCT correlate with the development of chronic GVHD
.
Bone Marrow Transplant.
2013
;
48
(
4
):
593
-
597
.
60.
Beri
N
,
Friedman
DR
,
Simms
TM
, et al
.
Molecular and clinical associations between vitamin D and chronic lymphocytic leukemia
.
Blood.
2013
;
122
(
21
):
5282
.
61.
Bhandari
R
,
Malvar
J
,
Sacapano
A
,
Aguayo-Hiraldo
P
,
Jodele
S
,
Orgel
E
.
Association between Vitamin D and risk for early and late post-transplant complications
.
Biol Blood Marrow Transplant.
2020
;
26
(
2
):
343
-
350
.
62.
Beebe
K
,
Magee
K
,
McNulty
A
, et al
.
Vitamin D deficiency and outcomes in pediatric hematopoietic stem cell transplantation
.
Pediatr Blood Cancer.
2018
;
65
(
2
):
e26817
.
63.
Wallace
G
,
Jodele
S
,
Howell
J
, et al
.
Vitamin D deficiency and survival in children after hematopoietic stem cell transplant
.
Biol Blood Marrow Transplant.
2015
;
21
(
9
):
1627
-
1631
.
64.
Perera
T
,
Lim
ABM
,
Mason
K
,
Szer
J
,
Ritchie
DS
.
The relationship between pre-transplant 25-hydroxy-vitamin D levels, survival and graft-versus-host disease, in allogeneic haematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant.
2015
;
21
(
2
):
S303
-
S304
.
65.
Campos
DJ
,
Biagini
GLK
,
Funke
VAM
,
Bonfim
CM
,
Boguszewski
CL
,
Borba
VZ
.
Vitamin D deficiency in children and adolescents submitted to hematopoietic stem cell transplantation
.
Rev Bras Hematol Hemoter.
2014
;
36
(
2
):
126
-
131
.
66.
Hansson
MEA
,
Norlin
AC
,
Omazic
B
, et al
.
Vitamin D levels affect outcome in pediatric hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant.
2014
;
20
(
10
):
1537
-
1543
.
67.
Beebe
K
,
Olsen
J
,
Chang
Y
, et al
.
Impact of vitamin D level pre and post allogeneic hematopoietic stem cell transplant
.
Blood.
2013
;
122
(
21
):
4616
.
68.
Sproat
L
,
Olsen
J
,
Beebe
K
, et al
.
Impact of Vitamin D level after allogeneic hematopoietic stem cell transplant
.
Blood.
2012
;
120
(
21
):
1954
.
69.
Hickish
T
,
Cunningham
D
,
Colston
K
, et al
.
The effect of 1,25-dihydroxyvitamin D3 on lymphoma cell lines and expression of vitamin D receptor in lymphoma
.
Br J Cancer.
1993
;
68
(
4
):
668
-
672
.
70.
Gascoyne
DM
,
Lyne
L
,
Spearman
H
,
Buffa
FM
,
Soilleux
EJ
,
Banham
AH
.
Vitamin D receptor expression in plasmablastic lymphoma and myeloma cells confers susceptibility to vitamin D
.
Endocrinology.
2017
;
158
(
3
):
503
-
515
.
71.
Pepper
C
,
Thomas
A
,
Hoy
T
,
Milligan
D
,
Bentley
P
,
Fegan
C
.
The vitamin D3 analog EB1089 induces apoptosis via a p53-independent mechanism involving p38 MAP kinase activation and suppression of ERK activity in B-cell chronic lymphocytic leukemia cells in vitro
.
Blood.
2003
;
101
(
7
):
2454
-
2460
.
72.
Bruns
H
,
Büttner
M
,
Fabri
M
, et al
.
Vitamin D-dependent induction of cathelicidin in human macrophages results in cytotoxicity against high-grade B cell lymphoma
.
Sci Transl Med.
2015
;
7
(
282
):
282ra47
.
73.
Kulling
PM
,
Olson
KC
,
Olson
TL
,
Feith
DJ
,
Loughran
TP
Jr
.
Vitamin D in hematological disorders and malignancies
.
Eur J Haematol.
2017
;
98
(
3
):
187
-
197
.
74.
Tao
Y
,
Chen
H
,
Zhou
Y
,
Shi
Y
.
Meta-analysis of the prognostic and clinical value of serum 25-hydroxyvitamin D levels in previously untreated lymphoma
.
Future Oncol.
2021
;
17
(
14
):
1825
-
1838
.
75.
Park
HY
,
Hong
YC
,
Lee
K
,
Koh
J
.
Vitamin D status and risk of non-Hodgkin lymphoma: an updated meta-analysis
.
PLoS One.
2019
;
14
(
4
):
e0216284
.
76.
Lu
D
,
Chen
J
,
Jin
J
.
Vitamin D status and risk of non-Hodgkin lymphoma: a meta-analysis
.
Cancer Causes Control.
2014
;
25
(
11
):
1553
-
1563
.
77.
Purdue
MP
,
Freedman
DM
,
Gapstur
SM
, et al
.
Circulating 25-hydroxyvitamin D and risk of non-Hodgkin lymphoma: Cohort Consortium Vitamin D Pooling Project of Rarer Cancers
.
Am J Epidemiol.
2010
;
172
(
1
):
58
-
69
.
78.
Rosenblatt
J
,
Bissonnette
A
,
Ahmad
R
, et al
.
Immunomodulatory effects of vitamin D: implications for GVHD
.
Bone Marrow Transplant.
2010
;
45
(
9
):
1463
-
1468
.
79.
Chiengthong
K
,
Cheungpasitporn
W
,
Thongprayoon
C
, et al
.
Vitamin D deficiency is not associated with graft versus host disease after hematopoietic stem cell transplantation: A meta-analysis
.
J Evid Based Med.
2020
;
13
(
3
):
183
-
191
.
80.
Vaughan-Shaw
PG
,
Buijs
LF
,
Blackmur
JP
, et al
.
The effect of vitamin D supplementation on survival in patients with colorectal cancer: systematic review and meta-analysis of randomised controlled trials
.
Br J Cancer.
2020
;
123
(
11
):
1705
-
1712
.
81.
Raoufinejad
K
,
Shamshiri
AR
,
Pezeshki
S
, et al
.
Oral calcitriol in hematopoietic recovery and survival after autologous stem cell transplantation: a randomized clinical trial
.
Daru.
2019
;
27
(
2
):
709
-
720
.
82.
Braegger
C
,
Campoy
C
,
Colomb
V
, et al;
ESPGHAN Committee on Nutrition
.
Vitamin D in the healthy European paediatric population
.
J Pediatr Gastroenterol Nutr.
2013
;
56
(
6
):
692
-
701
.
83.
Lips
P
.
Which circulating level of 25-hydroxyvitamin D is appropriate?
J Steroid Biochem Mol Biol.
2004
;
89-90
(
1-5
):
611
-
614
.
84.
Holick
MF
,
Binkley
NC
,
Bischoff-Ferrari
HA
, et al;
Endocrine Society
.
Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline
.
J Clin Endocrinol Metab.
2011
;
96
(
7
):
1911
-
1930
.
85.
Bischoff-Ferrari
HA
.
Optimal serum 25-hydroxyvitamin D levels for multiple health outcomes
.
Adv Exp Med Biol.
2008
;
624
:
55
-
71
.
86.
Zgaga
L
,
Theodoratou
E
,
Farrington
SM
, et al
.
Diet, environmental factors, and lifestyle underlie the high prevalence of vitamin D deficiency in healthy adults in Scotland, and supplementation reduces the proportion that are severely deficient
.
J Nutr.
2011
;
141
(
8
):
1535
-
1542
.
87.
Giustina
A
,
Adler
RA
,
Binkley
N
, et al
.
Controversies in vitamin D: summary statement from an international conference
.
J Clin Endocrinol Metab.
2019
;
104
(
2
):
234
-
240
.

Author notes

The full-text version of this article contains a data supplement.

Supplemental data