378
Views
1
CrossRef citations to date
0
Altmetric
Editorial

Post-transplant immunotherapy: combining cancer vaccines with hematopoietic stem cell transplantation

&
Pages 519-523 | Published online: 09 Jan 2014

The immune infiltrate is a significant part of the tumor micromillieu, and the quantity and quality of the infiltrate have clearly been linked to disease progression and patient survival Citation[1,2]. Correspondingly, immune-based targeting of cancer has been pursued for more than a century Citation[3] and prophylactic vaccination has already proven successful, at least against pathogen-induced cancers. Therefore, it is now routinely used for the prevention of human papilloma virus (HPV)-induced cervical cancer and HBV-induced liver cancer. These excellent examples could be employed as models for further obvious applications such as HPV-associated anal and penis carcinoma and EBV-associated post-transplant lymphoproliferative disease, Burkitt or Hodgkin’s lymphoma and even major cancers such as nasopharyngeal carcinoma in Asia (with an incidence of up to 30 out of 100,000 individuals affected by tumors) Citation[4].

However, therapeutic vaccination, although effective in murine models and often reported to induce tumor-specific T-cell responses in humans, has not yet experienced its clinical breakthrough. On the contrary, after more than two decades and over 1500 patients treated with dendritic cell (DC)-based vaccines (the major therapeutic strategy in cancer immunotherapy), overall response rates remain in the range of spontaneous regressions Citation[5]. Non-cell-based vaccines alike have been unable to meet the scientists’, physicians’ and – most importantly – patients’ expectations Citation[6].

This is surprising, as anticancer vaccination in mice has proven successful in many independent tumor models and immunologic settings. Furthermore, many tumor antigens and fundamental mechanisms of tumor evasion have been discovered and the knowledge has long entered the arena of immunotherapy Citation[7].

What is the difference between the successful examples previously mentioned and the disappointing reality in advanced-stage cancer patients? First, the choice of tumor antigen (foreign/mutated vs self-antigens) skews towards tumor control in prophylactic antiviral vaccinations in humans and many murine tumor models. Second, immune surveillance has been shown to correlate negatively with tumor load Citation[8]. The patients receiving anti-HBV or -HPV vaccinations and most of the murine models are free of tumor at the time of immunization and, thus, inhibitory mechanisms are not yet operative. Most importantly, the priming of an immune response against non-self antigens is an evolutionary necessity and, thus, much more powerful than the self-antigens chosen in many trials.

Therefore, the classic clinical scenario of advanced-stage patients with several kilograms of tumor burden that are vaccinated against self-antigens is a very challenging context indeed.

In this light, hematopoietic stem cell transplantation (HSCT) appears to be a unique setting for tumor vaccines: tumor load is generally required to be low before the patient is considered for transplantation and will further be reduced by the conditioning regimen; therefore, post-transplant levels of inhibitory cytokines and regulatory T cells (Tregs) appear to be low and there seems to be ‘space’ for the expansion of tumor-specific T cells. Furthermore, the context of allogeneic HSCT provides foreign antigens such as minor histocompatibility antigens (mHAGs) as promising targets.

Hence, what is the current status of anti-tumor vaccination in the HSCT setting and can these theoretical advantages be translated to a real therapeutic benefit?

Vaccination in the context of HSCT

In an ideal immunotherapist’s world, large numbers of highly reactive anti-tumor lymphocytes are generated in vivo without being restricted by tolerance mechanisms. The reconstituting immune system after HSCT provides a unique approximation to this ideal scenario. The lymphopenic environment allows strong expansion of respective anti-tumor T cells in the presence of cytokines responsible for thymic-independent homeostatic T-cell proliferation such as IL-7, IL-15 and IL-21 Citation[9]. In addition, pretransplant chemotherapy is known to be able to deplete regulatory cells Citation[10]. Hence, although there is profound post-transplant immunosuppression and especially a lack of support by helper T cells, there are ways to exploit this scenario to mount powerful CD8 responses Citation[11,12].

Against this background, both autologous and allogeneic strategies are attractive avenues to pursue specific immunologic targeting and lower the relapse rates of patients after HSCT. New developments in this field might indeed also revive transplantation for solid tumors Citation[13,14].

In autologous HSCT, unspecific immunostimulatory strategies used granulocyte–macrophage colony-stimulating factor (GM-CSF), IFN-γ or IL-2 (plus cyclosporine) to enhance the activity of reconstituting T cells and even mimic graft-versus-host (GvH) and graft-versus-tumor (GvT) effects known from allogeneic transplantation Citation[15,16]. Proof-of-principle studies demonstrated feasibility and treatment response.

Idiotype vaccination in the context of lymphoma and myeloma were among the first clinical vaccinations performed successfully, as comprehensively reviewed by de Cerio et al.Citation[17]. Idiotype vaccination was also tested following HSCT and specific anti-idiotype immune responses were demonstrated for lymphoma and myeloma patients Citation[18,19].

Intriguing autologous approaches have been introduced by groups from Stanford (CA, USA) and the NIH. The NIH group transferred autologous in vitro-expanded T cells derived from tumor-infiltrating lymphocytes (TILs) in combination with IL-2 to treat patients with metastatic melanoma after conditioning with total-body irradiation, fludarabine and cyclophosphamide. Objective response rates of 50–70% were achieved Citation[13]. This procedure is based on the successful expansion of CTLs recognizing relevant tumor-associated antigens as well as on the reduction of Tregs by radio/chemotherapy. In a further-developed experimental system, they were able to evade these limitations. Here, instead of using pre-existing TILs, genetically engineered peripheral blood lymphocytes carrying T-cell receptor chains specific for a melanoma antigen were transferred into patients. Sustained levels of these engineered cells could be observed in two patients with objective regression of metastatic melanoma lesions Citation[20].

The Stanford group initially injected CG-enriched oligonucleotides (CpGs) intratumorally after chemotherapeutic treatment, resulting in anti-tumor CD8 responses and the induction of remission in preclinical and clinical lymphoma models Citation[21]. In a murine system, they have now further developed this approach to enhance the anti-tumor effect by transferring tumor-specific CD8 lymphocytes from a vaccinated host into a syngeneic recipient followed by post-transplant booster vaccinations – a procedure they termed immunotransplantation Citation[22]. In a similar manner, Kochenderfer et al. induced vigorous CD8 responses after autologous transplantation using tumor peptide vaccines in conjunction with CpG and IL-2 Citation[23].

Allogeneic HSCT itself demonstrates the power of the immunologic approach to treat even cancers refractory to chemotherapy. The finding that mere infusion of donor lymphocytes (DLIs) could re-induce remission after relapse following allogeneic transplantation paved the way to exploit the GvT effect Citation[24]. Over the past decade, this strategy has been refined by a reduction of the chemotherapeutic aspect of the conditioning regimen prior to transplant. This shift towards the immunologic effect exerted by donor T cells allowed the extension of the indication for transplant and significantly decreased transplant-related mortality Citation[25]. Furthermore, it instigated numerous studies to improve the efficacy of the anti-tumor response through graft manipulation.

In general, anti-tumor activity can either be induced against unspecific allo-antigens or specific tumor- or leukemia-associated antigens (TAA/LAA). The efficacy of targeting TAA/LAA is especially well studied for epitopes derived from myeloid leukemia-associated antigens such as proteinase (PR)3, Wilms tumor (WT)-1 and BCR-ABL Citation[26]. Immunotherapeutic strategies either focus on the ex vivo generation of TAA/LAA-specific cytotoxic T cells (CTLs), which have been used successfully in relapse after allotransplantation Citation[27], or on using BCR-ABL, PR3 or WT1-targeted vaccines Citation[28].

In particular, alloreactive T cells evoke a strong immune response. Unfortunately, these alloantigen-induced immune responses (GvT responses) are often linked to the occurrence of GvH disease (GvHD) because tumor cells as well as the recipient’s healthy organ cells are detected as ‘foreign’. In this case, immunosuppressive therapy will have to be commenced or maintained, thereby diminishing the GvT effect.

Mismatched minor histocompatibility antigens present on tumor cells or only on cells of the hematopoietic system have been shown to induce GvT effects without significant GvHD Citation[29]. This strategy has now been taken to a new level by Goulmy’s group from Leiden who targeted the mHAG HA-1 by HA-1-mismatched CTLs and induced remissions in murine solid tumors aberrantly expressing HA-1 Citation[14]. In a second report, they were able to induce HA-1 expression in previously HA-1-negative tumors by the DNA-hypomethylating compound 5-aza-2´-deoxycytidine Citation[30], raising hopes that specific mHag-directed therapy could even revive allotransplantation in solid tumor patients.

Another strategy to benefit from a defined mismatch situation is to use donor-versus-recipient natural killer (NK) cell alloreactivity. Lack of inhibitory signals based on the absence of certain recipient HLA class I ligands for donor-inhibitory killer Ig-like receptors (KIRs) enable NK cells to eradicate leukemia cells as well as host antigen-presenting cells (APCs), thus reducing the incidence of GvHD Citation[31].

Donor lymphocyte infusion therapy has been a reasonably successful post-transplant immunotherapy since its discovery Citation[24]. However, response rates vary significantly depending on the disease and are generally rather low. There are different strategies to improve DLI efficacy. Ex vivo activation via CD3/CD28-coated beads improved DLI efficacy in chronic myeloid leukaemia patients without significantly increasing GvHD Citation[32]. A combination of DLI with antibodies or immunomodulatory agents such as interferon might increase responses and enable the reduction of DLI dosing to lower GvHD rates Citation[33,34]. The vaccination/priming of donors Citation[35,36] is another method that should be further evaluated.

Improving the response

All these immunotherapeutic strategies are based on intriguing ideas. However, translation into routine use will very much depend on their efficacy in clinical tumor regression.

Based on advancing immunologic knowledge, many groups work on improving the anti-tumor response with new strategies. Effectiveness of the Toll-like receptor 9 ligand CpG in the induction of vaccine responses was mentioned earlier. Eckl-Dorna et al. report how this CpG effect can now be directed to B cells via B-cell receptor-dependent uptake of CpG/antigens to induce antibody responses Citation[37]. In mice, CpGs also activate invariant NK T cells Citation[38], a subset of lipid-reactive innate lymphocytes previously shown to mount anti-tumor responses in humans after activation by CD1d-expressing DCs Citation[39,40].

Invariant NKT cell activation itself leads to the transactivation of NK T cells, another cell type of interest. In a murine system, these activated NK T cells help to turn so-called myeloid-derived suppressor cells into more immunogenic APCs Citation[41], boosting the CTL response and reducing Treg numbers.

Modification of peripheral tolerance mechanisms is also pursued by work on Treg depletion. A recombinant IL-2/diphtheria toxin conjugate (denileukin) has been successfully used to deplete FOXP3-positive Tregs and induce remission in metastases in mice and humans Citation[42,43]. Alternative methods to decrease Treg numbers or activity is to use gemcitabine Citation[44], fludarabine Citation[45] or cyclophosphamide chemotherapy Citation[46]. New transplant strategies omitting calcineurin inhibitors such as post-transplant cyclophosphamide as single-agent GvHD prophylaxis may allow an early expansion of CTLs and decrease the risk for relapse Citation[47].

Dendritic cells are often used as APCs to prime and expand tumor-specific T-cells in vivo or ex vivo. However, the generation of sufficient numbers of pure DCs for repetitive, potentially life-long vaccinations, as suggested in some murine models, is often not feasible and too expensive Citation[48]. Therefore, alternative strategies have been studied. Among these, CD40-activated B cells and artificial APCs appear to be a promising alternative, as they can be generated at the highest purity and in virtually unlimited amounts Citation[49,50]. Using antigen-loaded CD40-activated B cells to expand specific donor lymphocytes ex vivo prior to infusion followed by in vivo boosters is a strategy we find especially intriguing.

Taken together, HSCT appears to be an ideal scenario for immunotherapy in hematologic and maybe even solid tumors. However, successful and clinically feasible concepts need to be established. The identification of the best allo-antigens and the specific prevention of GvHD are among the major challenges that must be addressed. Nevertheless, current experimental strategies are promising and first clinical benefit may be seen in the near future.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Galon J, Costes A, Sanchez-Cabo F et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science313, 1960–1964 (2006).
  • Pages F, Berger A, Camus M et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N. Engl. J. Med.353, 2654–2666 (2005).
  • Coley WB. The treatment of malignant tumors by repeated inoculations of erysipelas, with a report of ten original cases. Am. J. Med. Sci.105, 487–511 (1893).
  • Titcomb CP Jr. High incidence of nasopharyngeal carcinoma in Asia. J. Insur. Med.33, 235–238 (2001).
  • Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat. Med.10, 909–915 (2004).
  • Fournier P, Schirrmacher V. Randomized clinical studies of anti-tumor vaccination: state of the art in 2008. Expert Rev. Vaccines8, 51–66 (2009).
  • Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat. Rev. Cancer5, 263–274 (2005).
  • Ochsenbein AF, Klenerman P, Karrer U et al. Immune surveillance against a solid tumor fails because of immunological ignorance. Proc. Natl Acad. Sci. USA96, 2233–2238 (1999).
  • Surh CD, Sprent J. Regulation of mature T cell homeostasis. Semin. Immunol.17, 183–191 (2005).
  • Symons HJ, Levy MY, Wang J et al. The allogeneic effect revisited: exogenous help for endogenous, tumor-specific T cells. Biol. Blood Marrow Transplant.14, 499–509 (2008).
  • Bandera A, Trabattoni D, Pacei M et al. Fully immunocompetent CD8+ T lymphocytes are present in autologous haematopoietic stem cell transplantation recipients despite an ineffectual T-helper response. PLoS ONE3, e3616 (2008).
  • Rapoport AP, Stadtmauer EA, Aqui N et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat. Med.11, 1230–1237 (2005).
  • Dudley ME, Yang JC, Sherry R et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J. Clin. Oncol.26, 5233–5239 (2008).
  • Hambach L, Vermeij M, Buser A et al. Targeting a single mismatched minor histocompatibility antigen with tumor-restricted expression eradicates human solid tumors. Blood112, 1844–1852 (2008).
  • Bolanos-Meade J, Garrett-Mayer E, Luznik L et al. Induction of autologous graft-versus-host disease: results of a randomized prospective clinical trial in patients with poor risk lymphoma. Biol. Blood Marrow Transplant.13, 1185–1191 (2007).
  • Stein AS, O’Donnell MR, Slovak ML et al. Interleukin-2 after autologous stem-cell transplantation for adult patients with acute myeloid leukemia in first complete remission. J. Clin. Oncol.21, 615–623 (2003).
  • de Cerio AL, Inoges S. Future of idiotypic vaccination for B-cell lymphoma. Expert Rev. Vaccines8, 43–50 (2009).
  • Bendandi M, Rodriguez-Calvillo M, Inoges S et al. Combined vaccination with idiotype-pulsed allogeneic dendritic cells and soluble protein idiotype for multiple myeloma patients relapsing after reduced-intensity conditioning allogeneic stem cell transplantation. Leuk. Lymphoma47, 29–37 (2006).
  • Davis TA, Hsu FJ, Caspar CB et al. Idiotype vaccination following ABMT can stimulate specific anti-idiotype immune responses in patients with B-cell lymphoma. Biol. Blood Marrow Transplant.7, 517–522 (2001).
  • Morgan RA, Dudley ME, Wunderlich JR et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science314, 126–129 (2006).
  • Li J, Song W, Czerwinski DK et al. Lymphoma immunotherapy with CpG oligodeoxynucleotides requires TLR9 either in the host or in the tumor itself. J. Immunol.179, 2493–2500 (2007).
  • Brody JD, Goldstein MJ, Czerwinski DK, Levy R. Immunotransplantation preferentially expands T-effector cells over T-regulatory cells and cures large lymphoma tumors. Blood113, 85–94 (2009).
  • Kochenderfer JN, Simpson JL, Chien CD, Gress RE. Vaccination regimens incorporating CpG-containing oligodeoxynucleotides and IL-2 generate antigen-specific antitumor immunity from T-cell populations undergoing homeostatic peripheral expansion after BMT. Blood110, 450–460 (2007).
  • Kolb HJ, Mittermuller J, Clemm C et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood76, 2462–2465 (1990).
  • Copelan EA. Hematopoietic stem-cell transplantation. N. Engl. J. Med.354, 1813–1826 (2006).
  • Rezvani K, Barrett AJ. Characterizing and optimizing immune responses to leukaemia antigens after allogeneic stem cell transplantation. Best Pract. Res. Clin. Haematol.21, 437–453 (2008).
  • Marijt E, Wafelman A, van der Hoorn M et al. Phase I/II feasibility study evaluating the generation of leukemia-reactive cytotoxic T lymphocyte lines for treatment of patients with relapsed leukemia after allogeneic stem cell transplantation. Haematologica92, 72–80 (2007).
  • Rezvani K, Yong AS, Mielke S et al. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood111, 236–242 (2008).
  • Marijt WA, Heemskerk MH, Kloosterboer FM et al. Hematopoiesis-restricted minor histocompatibility antigens HA-1- or HA-2-specific T cells can induce complete remissions of relapsed leukemia. Proc. Natl Acad. Sci. USA100, 2742–2747 (2003).
  • Hambach L, Ling KW, Pool J et al. Hypomethylating drugs convert HA-1 negative solid tumors into targets for stem cell based immunotherapy. Blood113(12), 2715–2722 (2008).
  • Ruggeri L, Capanni M, Urbani E et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science295, 2097–2100 (2002).
  • Porter DL, Levine BL, Bunin N et al. A Phase 1 trial of donor lymphocyte infusions expanded and activated ex vivo via CD3/CD28 costimulation. Blood107, 1325–1331 (2006).
  • Buhmann R, Simoes B, Stanglmaier M et al. Immunotherapy of recurrent B-cell malignancies after allo-SCT with Bi20 (FBTA05), a trifunctional anti-CD3 × anti-CD20 antibody and donor lymphocyte infusion. Bone Marrow Transplant43(5), 383–397 (2008).
  • Posthuma EF, Marijt EW, Barge RM et al. α-interferon with very-low-dose donor lymphocyte infusion for hematologic or cytogenetic relapse of chronic myeloid leukemia induces rapid and durable complete remissions and is associated with acceptable graft-versus-host disease. Biol. Blood Marrow Transplant.10, 204–212 (2004).
  • Cabrera R, Diaz-Espada F, Barrios Y et al. Infusion of lymphocytes obtained from a donor immunised with the paraprotein idiotype as a treatment in a relapsed myeloma. Bone Marrow Transplant25, 1105–1108 (2000).
  • Neelapu SS, Munshi NC, Jagannath S et al. Tumor antigen immunization of sibling stem cell transplant donors in multiple myeloma. Bone Marrow Transplant36, 315–323 (2005).
  • Eckl-Dorna J, Batista FD. BCR-mediated uptake of antigen linked to TLR9-ligand stimulates B-cell proliferation and antigen-specific plasma cell formation. Blood DOI: 0: blood-2008-10-185421v1 (2009) (Epub ahead of print).
  • Paget C, Bialecki E, Fontaine J et al. Role of invariant NK T lymphocytes in immune responses to CpG oligodeoxynucleotides. J. Immunol.182, 1846–1853 (2009).
  • Nieda M, Okai M, Tazbirkova A et al. Therapeutic activation of Vα24+Vβ11+ NKT cells in human subjects results in highly coordinated secondary activation of acquired and innate immunity. Blood103, 383–389 (2004).
  • Ishikawa A, Motohashi S, Ishikawa E et al. A Phase I study of α-galactosylceramide (KRN7000)-pulsed dendritic cells in patients with advanced and recurrent non-small cell lung cancer. Clin. Cancer Res.11, 1910–1917 (2005).
  • Ko HJ, Lee JM, Kim YJ et al. Immunosuppressive myeloid-derived suppressor cells can be converted into immunogenic APCs with the help of activated NKT cells: an alternative cell-based antitumor vaccine. J. Immunol.182, 1818–1828 (2009).
  • Morse MA, Hobeika AC, Osada T et al. Depletion of human regulatory T cells specifically enhances antigen-specific immune responses to cancer vaccines. Blood112, 610–618 (2008).
  • Dannull J, Su Z, Rizzieri D et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J. Clin. Invest.115, 3623–3633 (2005).
  • Correale P, Tagliaferri P, Fioravanti A et al. Immunity feedback and clinical outcome in colon cancer patients undergoing chemoimmunotherapy with gemcitabine + FOLFOX followed by subcutaneous granulocyte macrophage colony-stimulating factor and aldesleukin (GOLFIG-1 Trial). Clin. Cancer. Res.14, 4192–4199 (2008).
  • Beyer M, Kochanek M, Darabi K et al. Reduced frequencies and suppressive function of CD4+CD25hi regulatory T cells in patients with chronic lymphocytic leukemia after therapy with fludarabine. Blood106, 2018–2025 (2005).
  • Kottke T, Thompson J, Diaz RM et al. Improved systemic delivery of oncolytic reovirus to established tumors using preconditioning with cyclophosphamide-mediated Treg modulation and interleukin-2. Clin. Cancer Res.15, 561–569 (2009).
  • Luznik L, Fuchs EJ, Chen AR et al. Post-translpantation high-dose cyclophosphamide (CY) is effective single agent GvHD prophylaxis that permits prompt immune reconstitution after myeloablative HLA matched related and unrelated bone marrow transplantation (BMT). Biol. Blood Marrow Transplant.13, 4 (2007).
  • Figdor CG, de Vries IJ, Lesterhuis WJ, Melief CJ. Dendritic cell immunotherapy: mapping the way. Nat. Med.10, 475–480 (2004).
  • Oelke M, Maus MV, Didiano D et al.Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig-coated artificial antigen-presenting cells. Nat. Med.9, 619–624 (2003).
  • von Bergwelt-Baildon MS, Vonderheide RH, Maecker B et al. Human primary and memory cytotoxic T lymphocyte responses are efficiently induced by means of CD40-activated B cells as antigen-presenting cells: potential for clinical application. Blood99, 3319–3325 (2002).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.