Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Sep 19.
Published in final edited form as: Am J Hematol. 2011 Oct;86(10):879–882. doi: 10.1002/ajh.22136

Peri-transplant palifermin use and lymphocyte recovery after T-cell replete, matched related allogeneic hematopoietic cell transplantation

Romee Rizwan 1, John E Levine 2, Todd DeFor 1, James LM Ferarra 2, Daniel J Weisdorf 1, Bruce R Blazar, Michael R Verneris 1
PMCID: PMC3446845  NIHMSID: NIHMS404459  PMID: 21922528

Abstract

A previously conducted randomized, double-blind, placebo-controlled study from 2000-2003 tested peri-transplant palifermin (KGF) in 100 recipients of T-replete matched related donor allogeneic hematopoietic transplant (MRD allo-HCT). The use of KGF in preclinical transplant models, including an autologous non-human primate model, has been associated with improved immune reconstitution. Therefore, we investigated whether palifermin treated patients (n = 69) had improved absolute lymphocyte counts (ALCs) at days 30, 60 and 100 after transplant compared to placebo treated patients (n = 31). No statistically significant difference in ALC was noted at these time points. Additionally, there was no difference in ALC between patients who received low (240μg/kg) vs. high (720μg/kg) dose palifermin. Patients with a day 30 ALC of >600 ×106/L had a trend towards improved progression-free survival (49% vs. 29%, p=0.07), lower transplant-related mortality (18% vs. 35%, p=0.1) and grade II-IV aGvHD (31% vs. 47%, p=0.14), but this was not influenced by palifermin therapy. We conclude that following myeloablation and a T-replete MSD allogeneic graft, peri-transplant palifermin does not accelerate early lymphocyte recovery. Studies combining palifermin with other agents (as in pre-clinical models) may be required to improve immune reconstitution after allo-HCT.

Keywords: KGF, absolute lymphocyte count, allogeneic hematopoietic cell transplantation, myeloablative conditioning


Allogeneic hematopoietic cell transplantation (allo-HCT) is often the only curative option for people with otherwise fatal hematologic malignancies. As the number of allo-HCT procedures continues to increase [1], it is increasingly clear that a major obstacle to success is delayed immune recovery, which puts atients at risk for a wide variety of opportunistic infections [2-7][8]. Additionally, rapid early lymphocyte recovery may serve as a surrogate predictor of better transplant outcomes. Robust recovery of absolute lymphocyte counts (ALC) early after transplantation is associated with improved survival following autologous, sibling, unrelated bone marrow, peripheral blood and umbilical cord blood transplantation [9-15]. There is a clear need to develop strategies to accelerate and improve immune reconstitution (IR). Several novel approaches have been successfully tested in preclinical animal models and early human clinical trials. These include pre-transplant androgen ablation, keratinocyte growth factor (KGF), and a p53 inhibitor or post-transplant administration of IL-7, IL-15, growth hormone, or insulin-like growth factor-1 [16-20].

KGF is a potent growth factor belonging to the fibroblast growth factor family that stimulates epithelial growth and differentiation, without affecting the non-epithelial cells that lack the FGFR2-IIIb receptor. In the thymus KGF is produced locally by mesenchymal cells and acts on thymic epithelial cells (TECs) [18, 21]. Through its trophic effects on TECs, KGF facilitates normal thymopoiesis which is required for naïve T cell generation [22]. In murine allo-HCT models, KGF treatment improved thymopoiesis and enhanced T cell IR [18, 23]. In a nonhuman primate model of autologous hematopoietic cell rescue following myeloablation, peri-transplant KGF administration led to the preservation of thymic architecture for up to 12 months after HCT [24]. In the same study, KGF treated animals had higher frequencies of naïve T cells compared to the control group. Another preclinical study which combined KGF with androgen depletion led to enhanced thymopoiesis characterized by a broad V-beta T cell repertoire [25].

To assess the GvHD protective potential of KGF, a phase I/II randomized, placebo controlled trial of recombinant human KGF (palifermin) was performed from 2000-2003 in 100 patients undergoing T cell replete, MRD allo-HCT following myeloablative conditioning [26, 27]. The focus of this study was to determine the impact of KGF on GVHD. Detailed IR was not studied within the trial. Because ALC is associated with transplant outcomes [9-15]. we questioned whether there might be differential effects of KGF on early ALC recovery. Here, we report our retrospective analysis using data from our earlier trial to assess the effects of peritransplant use of palifermin on ALC recovery at following HCT. This is the first report analyzing the results of palifermin and lymphocyte recovery in humans.

We tested ALCs after transplant in the palifermin and placebo treatment groups. As shown in figure 1a, the median ALC at days 30, 60 and 100 after transplant did not significantly differ in the palifermin vs. placebo arms (600 × 106/L vs 600 × 106/L; P = 0.26, 500 × 106/L vs 600 × 106/L; P = 0.86, 600 × 106/L vs 700 × 106/L, P = 0.19). Likewise, there was no influence of the palifermin dose and subsequent ALC recovery post-transplant ALC (figure 1b). As well, palifermin did not impact ALC based on the conditioning regimen (Bu/Cy vs. Cy/TBI), recipient age and the presence of aGVHD (data not shown). As previously reported, in this trial, palifermin use was not associated with shorter time to engraftment, aGvHD, survival or infectious complications after transplant. However, palifermin modestly decreased mucositis [26, 27].

Figure 1. Post-transplant ALC in palifermin and placebo groups.

Figure 1

A) Median and inter-quartile range of ALC for the placebo (open bars) and palifermin (shaded bars) at day 30, 60 and 100 after transplantation. The median ALC at the three time points in the two groups were 600 × 106/L vs. 600 × 106/L (P = 0.26), 600 × 106/L vs. 500 × 106/L (P = 0.86) and 700 × 106/L vs. 600 × 106/L (P = 0.19), respectively. B) Comparison of median and inter-quartile ranges of day 30, 60 and 90 ALCs at various doses of palifermin with placebo.

Based on prior studies [9-15], we also examined whether differential lymphocyte counts were associated with transplant outcomes. Patients with an ALC above the medin at D+30 (>600 ×106/L) showed a trend for improved PFS (49% (95% CI, 32-65%) vs. 29% (95% CI 16-43%), p=0.07) (figure 2a). There was no association between D+30 ALC and disease recurrence (28% (95% CI 14-42%) vs. 23% (95% CI 10-36%), p=0.6) (figure 2b). There were trends toward less 2 year TRM (95% CI 18% (6-30%) vs. 35% (95% CI 20-50%), p=0.1) and grade II-IV aGVHD (31% (95% CI 17-45%) vs. 47% (95% CI 32-62%, p=0.14) in patients with an ALC>600 at D+30 after transplant (figures 2c and 2d). These rates were unaffected by KGF therapy.

Figure 2. Transplant outcomes based on ALC at day 30.

Figure 2

Outcomes were measured for patients above or below the median ALC (i.e, >600 × 106/L or ≤ 600) at day 30. A) PFS at Day 30 was 49% vs. 29%, respectively (p=0.07). B) The cumulative incidence of relapse was 28% vs. 23%, respectively (p=0.6). C) 2-year TRM was 18% vs. 35%, respectively (p=0.1). D) Grade II-IV aGvHD was 31 % vs. 47%, (p=0.14).

In this first allotransplant palifermin dose escalation trial, using ALC as a marker for IR, we found that peri-transplant palifermin had no impact on ALC at day 30, 60 or 100 after allo-HCT. These results differ from rodent studies [22, 25] and a non-human primate study [24] where KGF was associated with protection of TECs from radiation-induced damage, resulting in improved thymopoiesis and peripheral IR after transplant. Numerous explanations may account for our the findings including suboptimal palifermin dose or dosing schedule, existing pre-transplant chemotherapy-induced TEC damage or a limited potency of this agent on human TECs. ALC is a relatively crude marker for IR and the lack of information on various T cell subsets, T cell receptor Vβ repertoire and newly formed T cells (i.e., thymic excision circles) limits our ability to detect any effects of palifermin on the naïve vs. homeostatically expanded T cells. In addition, only three patients were less than 18 years in this study, with the median age being 47. The lack of accelerated IR and higher ALC with palifermin could also be related to a reduced effectiveness of palifermin on atrophied thymic tissue, present in older subjects. Alternatively, a recent study by Chakraverty et al found that the degree of in vivo T cell depletion adversely affects ALC recovery post allo-HCT, underscoring the importance of graft-derived T cells in early ALC recovery [29]. Therefore it is possible that the early lymphocyte recovery after T cell replete HCT is not reflective of thymic output and may be more related to homeostatic proliferation of existing T cells. In this scenario, ALC may be unaffected by palifermin. In addition, significant aGVHD occurred in nearly half the patients on this study. This complication is associated with thymic injury and requires extended immunosuppressive therapy. Thus aGVHD (or its treatment) might have confounded any potential of palifermin to augment IR. However, there was no significant difference in ALCs between the palifermin and placebo treated groups, even after excluding patients with aGvHD (data not shown). However it is still possible that the GvHD prophylaxis itself (which were not used in the preclinical models) may have blunted any potential benefit of palifermin on lymphocyte recovery, considering that most GVHD prophylactic agents inhibit T cell production and expansion.

Previous studies show a positive correlation between early ALC and transplant with outcome measures [9, 14, 30]. While the clinical impact of ALC recovery was only a secondary aim of this study, we observed a trend towards improved PFS and lower TRM in patients above the median ALC at day 30, consistent with other reports [13-15, 30]. Interestingly, a lower day 30 ALC was associated with a trend towards more frequent grade II-IV aGvHD. Thus, it is possible that before becoming clinically evident, aGvHD is heralded by delayed recovery of ALC.

Delayed IR continues to be a major problem after allo-HCT. Based on this analysis, palifermin alone is unlikely to significantly improve post allo-HCT immune recovery, at least following T-cell replete allo-HCT. In recent years there have been significant advances in understanding the mechanisms behind delayed immune reconstitution and some novel interventions have been successful in pre-clinical models (reviewed in [28]). Perhaps strategies combining two or more agents, shown to be promising in pre-clinical models, may be needed to overcome the profound immune deficiency which follows allo-HCT.

Patients and methods

Patient and transplant characteristics

Patient and transplant characteristics have been reported previously [26, 27]. 100 patients were enrolled in this study and randomized to receive placebo (n=31) or palifermin (n=69). Two patients in the palifermin group did not undergo transplantation and were excluded. As shown in table 1, patient demographics and underlying diagnosis were well matched between the two groups. Median follow-up among survivors is 4.1 years (range: 2-6.1 years) for both groups.

Table 1.

Patient baseline demographics and disease characteristics

N Palifermin Placebo P

Center 0.91
    Michigan 46 32 14
    Minnesota 54 37 17

Gender 0.99
    Male 58 40 18
    Female 42 29 13

Median Age, y (range) 46 (7-65) 46 (7-63) 0.58

Diagnosis 0.08
    AML 36 24 12
    CML 15 7 8
    MDS 12 6 6
    NHL 14 13 1
    ALL 9 8 1
    Hodgkin's disease 1 1 0
    Other malignancies 13 10 3

AML indicates acute myelogenous leukemia; CML, chronic myelogenous leukemia; MDS, myelodysplasia syndrome; NHL, non-Hodgkin lymphoma.

All patients received allo-HCT from HLA-identical sibling donors after myeloablative conditioning which included 120 mg of cyclophosphamide and 1320 cGy of fractionated total body irradiation (TBI) at University of Minnesota or 16 mg/kg of oral busulfan and 60 mg/kg of cyclophosphamide (Bu/Cy) at the University of Michigan. A calcineurin inhibitor (cyclosporine or tacrolimus) and methotrexate (15 mg/m2 on day 1 and 10 mg/m2 on days 3, 6 and 11) were used for GvHD prophylaxis.

Study design

This was a randomized, double-blind, placebo-controlled, dose-escalation study. Three cohorts receiving increasing palifermin doses were sequentially enrolled, with each cohort randomized to achieve balance within each study site and in each cohort. Stratification was based on conditioning regimen and patient age. A total of 98 patients completed this study, with a 1:2 randomization between placebo (n=31) and study drug (n=67). All treatment cohorts received 3 days of palifermin at 40 mg/kg (n=8) or 60 mg/kg (n=61) prior to the start of conditioning (days -11 to -9) and then received the same palifermin dose on 3 consecutive days weekly (first three days of a week) staring on day 0 and extending for 1 (n=18), 2 (n=14) or 3 (n=37) weeks after transplant. Thus, the total palifermin dose was 240 mg/kg in the lowest dose cohort and 720 mg/kg for the highest dose cohort. For the current analysis, absolute lymphocyte counts (ALCs) were extracted from the patient medical records on days 30, 60 and 100 (+/- 7 days) after obtaining institutional review board approvals at both study sites.

Statistical analysis

Statistical comparisons of the distribution of ALC over time were performed by examining the median, inter-quartile ranges and ranges of ALC at days 30, 60 and 100 post transplant and performing either a general Wilcoxon or Kruskal-Wallis test by a number of factors: KGF group (placebo versus KGF), placebo dose, presence of grade II-IV acute GvHD, conditioning and age.

Outcomes by ALC were analyzed by simply dividing cohorts by the median ALC and estimating the outcomes of the two groups using Kaplan-Meier estimates for overall survival (OS) and progression-free survival (PFS) (34) and the cumulative incidence (CI) for relapse, non-relapse mortality (NRM) and GvHD (35). Analyses were performed using SAS 9.2 (SAS Institute) and R 2.4 statistical software.

Acknowledgments

This work was supported in part by grants from the National Institutes of Health NCI P01-CA65493 (BRB, and MRV), American Cancer Society RSG-08-181-LIB (MRV), Leukemia Research Fund (MRV) and Children's Cancer Research Fund (MRV and BRB).

Footnotes

Author Contribution

Romee Rizwan: planned data analysis, reviewed and complied data, wrote paper

Todd DeFor: performed statistical analysis

John Levine: planned the original trial, provided data and edited the manuscript

James Ferarra: planned the original clinical trial and edited the manuscript

Daniel Weisdorf: planned the original clinical trial and edited the manuscript

Bruce R. Blazar: planned the original clinical trial and edited the manuscript

Michael R. Verneris: planned data analysis, reviewed and complied data, wrote paper

Reference

  • 1.Ballen KK, King RJ, Chitphakdithai P, et al. The national marrow donor program 20 years of unrelated donor hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2008;14:2–7. doi: 10.1016/j.bbmt.2008.05.017. [DOI] [PubMed] [Google Scholar]
  • 2.Shilling HG, McQueen KL, Cheng NW, et al. Reconstitution of NK cell receptor repertoire following HLA-matched hematopoietic cell transplantation. Blood. 2003;101:3730–3740. doi: 10.1182/blood-2002-08-2568. [DOI] [PubMed] [Google Scholar]
  • 3.Triplett BM, Horwitz EM, Iyengar R, et al. Effects of activating NK cell receptor expression and NK cell reconstitution on the outcomes of unrelated donor hematopoietic cell transplantation for hematologic malignancies. Leukemia. 2009;23:1278–1287. doi: 10.1038/leu.2009.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Douek DC, Vescio RA, Betts MR, et al. Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution. Lancet. 2000;355:1875–1881. doi: 10.1016/S0140-6736(00)02293-5. [DOI] [PubMed] [Google Scholar]
  • 5.Heitger A, Neu N, Kern H, et al. Essential role of the thymus to reconstitute naive (CD45RA+) T-helper cells after human allogeneic bone marrow transplantation. Blood. 1997;90:850–857. [PubMed] [Google Scholar]
  • 6.Marie-Cardine A, Divay F, Dutot I, et al. Transitional B cells in humans: characterization and insight from B lymphocyte reconstitution after hematopoietic stem cell transplantation. Clin Immunol. 2008;127:14–25. doi: 10.1016/j.clim.2007.11.013. [DOI] [PubMed] [Google Scholar]
  • 7.Cooley S, McCullar V, Wangen R, et al. KIR reconstitution is altered by T cells in the graft and correlates with clinical outcomes after unrelated donor transplantation. Blood. 2005;106:4370–4376. doi: 10.1182/blood-2005-04-1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Szabolcs P, Niedzwiecki D. Immune reconstitution after unrelated cord blood transplantation. Cytotherapy. 2007;9:111–122. doi: 10.1016/j.bbmt.2007.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Burke MJ, Vogel RI, Janardan SK, et al. Early Lymphocyte Recovery and Outcomes after Umbilical Cord Blood Transplantation (UCBT) for Hematologic Malignancies. Biol Blood Marrow Transplant. 2010 doi: 10.1016/j.bbmt.2010.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pavletic ZS, Joshi SS, Pirruccello SJ, et al. Lymphocyte reconstitution after allogeneic blood stem cell transplantation for hematologic malignancies. Bone Marrow Transplant. 1998;21:33–41. doi: 10.1038/sj.bmt.1701037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Le Blanc K, Barrett AJ, Schaffer M, et al. Lymphocyte recovery is a major determinant of outcome after matched unrelated myeloablative transplantation for myelogenous malignancies. Biol Blood Marrow Transplant. 2009;15:1108–1115. doi: 10.1016/j.bbmt.2009.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kumar S, Chen MG, Gastineau DA, et al. Lymphocyte recovery after allogeneic bone marrow transplantation predicts risk of relapse in acute lymphoblastic leukemia. Leukemia. 2003;17:1865–1870. doi: 10.1038/sj.leu.2403055. [DOI] [PubMed] [Google Scholar]
  • 13.Kumar S, Chen MG, Gastineau DA, et al. Effect of slow lymphocyte recovery and type of graft-versus-host disease prophylaxis on relapse after allogeneic bone marrow transplantation for acute myelogenous leukemia. Bone Marrow Transplant. 2001;28:951–956. doi: 10.1038/sj.bmt.1703262. [DOI] [PubMed] [Google Scholar]
  • 14.Ishaqi MK, Afzal S, Dupuis A, et al. Early lymphocyte recovery post- allogeneic hematopoietic stem cell transplantation is associated with significant graft-versus-leukemia effect without increase in graft-versus-host disease in pediatric acute lymphoblastic leukemia. Bone Marrow Transplant. 2008;41:245–252. doi: 10.1038/sj.bmt.1705891. [DOI] [PubMed] [Google Scholar]
  • 15.Kim DH, Kim JG, Sohn SK, et al. Clinical impact of early absolute lymphocyte count after allogeneic stem cell transplantation. Br J Haematol. 2004;125:217–224. doi: 10.1111/j.1365-2141.2004.04891.x. [DOI] [PubMed] [Google Scholar]
  • 16.Kelly RM, Goren EM, Taylor PA, et al. Short-term inhibition of p53 combined with keratinocyte growth factor improves thymic epithelial cell recovery and enhances T-cell reconstitution after murine bone marrow transplantation. Blood. 2010;115:1088–1097. doi: 10.1182/blood-2009-05-223198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rossi SW, Jeker LT, Ueno T, et al. Keratinocyte growth factor (KGF) enhances postnatal T-cell development via enhancements in proliferation and function of thymic epithelial cells. Blood. 2007;109:3803–3811. doi: 10.1182/blood-2006-10-049767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Min D, Taylor PA, Panoskaltsis-Mortari A, et al. Protection from thymic epithelial cell injury by keratinocyte growth factor: a new approach to improve thymic and peripheral T-cell reconstitution after bone marrow transplantation. Blood. 2002;99:4592–4600. doi: 10.1182/blood.v99.12.4592. [DOI] [PubMed] [Google Scholar]
  • 19.Min D, Panoskaltsis-Mortari A, Kuro OM, et al. Sustained thymopoiesis and improvement in functional immunity induced by exogenous KGF administration in murine models of aging. Blood. 2007;109:2529–2537. doi: 10.1182/blood-2006-08-043794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Napolitano LA, Schmidt D, Gotway MB, et al. Growth hormone enhances thymic function in HIV-1-infected adults. J Clin Invest. 2008;118:1085–1098. doi: 10.1172/JCI32830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Erickson M, Morkowski S, Lehar S, et al. Regulation of thymic epithelium by keratinocyte growth factor. Blood. 2002;100:3269–3278. doi: 10.1182/blood-2002-04-1036. [DOI] [PubMed] [Google Scholar]
  • 22.Alpdogan O, Hubbard VM, Smith OM, et al. Keratinocyte growth factor (KGF) is required for postnatal thymic regeneration. Blood. 2006;107:2453–2460. doi: 10.1182/blood-2005-07-2831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rossi S, Blazar BR, Farrell CL, et al. Keratinocyte growth factor preserves normal thymopoiesis and thymic microenvironment during experimental graft-versus-host disease. Blood. 2002;100:682–691. doi: 10.1182/blood.v100.2.682. [DOI] [PubMed] [Google Scholar]
  • 24.Seggewiss R, Lore K, Guenaga FJ, et al. Keratinocyte growth factor augments immune reconstitution after autologous hematopoietic progenitor cell transplantation in rhesus macaques. Blood. 2007;110:441–449. doi: 10.1182/blood-2006-12-065623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kelly RM, Highfill SL, Panoskaltsis-Mortari A, et al. Keratinocyte growth factor and androgen blockade work in concert to protect against conditioning regimen-induced thymic epithelial damage and enhance T-cell reconstitution after murine bone marrow transplantation. Blood. 2008;111:5734–5744. doi: 10.1182/blood-2008-01-136531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Blazar BR, Weisdorf DJ, Defor T, et al. Phase 1/2 randomized, placebo-control trial of palifermin to prevent graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT). Blood. 2006;108:3216–3222. doi: 10.1182/blood-2006-04-017780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Levine JE, Blazar BR, DeFor T, et al. Long-term follow-up of a phase I/II randomized, placebo-controlled trial of palifermin to prevent graft-versus-host disease (GVHD) after related donor allogeneic hematopoietic cell transplantation (HCT). Biol Blood Marrow Transplant. 2008;14:1017–1021. doi: 10.1016/j.bbmt.2008.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Seggewiss R, Einsele H. Immune reconstitution after allogeneic transplantation and expanding options for immunomodulation: an update. Blood. 2010;115:3861–3868. doi: 10.1182/blood-2009-12-234096. [DOI] [PubMed] [Google Scholar]
  • 29.Chakraverty R, Orti G, Roughton M, et al. Impact of in vivo alemtuzumab dose before reduced intensity conditioning and HLA-identical sibling stem cell transplantation: pharmacokinetics, GVHD, and immune reconstitution. Blood. 2010;116:3080–3088. doi: 10.1182/blood-2010-05-286856. [DOI] [PubMed] [Google Scholar]
  • 30.Savani BN, Mielke S, Rezvani K, et al. Absolute lymphocyte count on day 30 is a surrogate for robust hematopoietic recovery and strongly predicts outcome after T cell-depleted allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2007;13:1216–1223. doi: 10.1016/j.bbmt.2007.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES