371
Views
0
CrossRef citations to date
0
Altmetric
Reviews

Targeting the immune microenvironment in mantle cell lymphoma: implications for current and emerging therapies

, &
Pages 2515-2527 | Received 16 Feb 2022, Accepted 27 May 2022, Published online: 15 Jun 2022

References

  • Eskelund CW, Dahl C, Hansen JW, et al. TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood J Am Soc Hematol. 2017;130(17):1903–1910.
  • Kumar A, Soumerai JD, Abramson JS, et al. Preliminary safety and efficacy from a multicenter, investigator-initiated phase ii study in untreated tp53 mutant mantle cell lymphoma with zanubrutinib, obinutuzumab, and venetoclax (BOVen). Blood. 2021;138(Supplement 1):3540–3540.
  • Papin A, Gouill SL, Chiron D. Rationale for targeting tumor cells in their microenvironment for mantle cell lymphoma treatment. Leuk Lymphoma. 2018;59(5):1064–1072.
  • MCL Cells in Tumor Microenvironment Impair T-Cell Metabolic Fitness and Effector Function. ASH; 2020.
  • Balsas P, Veloza L, Clot G, et al. SOX11, CD70 and Treg cells configure the tumor immune microenvironment of aggressive mantle cell lymphoma. Blood. 2021;138(22):2202–2215.
  • Yang ZZ, Novak AJ, Ziesmer SC, et al. CD70+ non-Hodgkin lymphoma B cells induce Foxp3 expression and regulatory function in intratumoral CD4 + CD25− T cells. Blood Am Soc Hematol. 2007;110(7):2537–2544.
  • Le K, Sun J, Khawaja H, et al. Mantle cell lymphoma polarizes tumor-associated macrophages into M2-like macrophages, which in turn promote tumorigenesis. Blood Adv. 2021;5(14):2863–2863.
  • Medina DJ, Goodell L, Glod J, et al. Mesenchymal stromal cells protect mantle cell lymphoma cells from spontaneous and drug-induced apoptosis through secretion of B-cell activating factor and activation of the canonical and non-canonical nuclear factor κB pathways. Haematologica. 2012;97(8):1255–1255.
  • Kurtova AV, Tamayo AT, Ford RJ, et al. Mantle cell lymphoma cells express high levels of CXCR4, CXCR5, and VLA-4 (CD49d): importance for interactions with the stromal microenvironment and specific targeting. Blood. 2009;113(19):4604–4604.
  • Zhang L, Yang J, Qian J, et al. Role of the microenvironment in mantle cell lymphoma: IL-6 is an important survival factor for the tumor cells. Blood. 2012;120(18):3783–3783.
  • Chiron D, Bellanger C, Papin A, et al. Rational targeted therapies to overcome microenvironment-dependent expansion of mantle cell lymphoma. Blood. 2016;128(24):2808–2818.
  • Zhang S, Jiang VC, Han G, et al. Longitudinal single-cell profiling reveals molecular heterogeneity and tumor-immune evolution in refractory mantle cell lymphoma. Nat Commun. 2021;12(1):1–17.
  • Wang L, Qian J, Lu Y, et al. Immune evasion of mantle cell lymphoma: expression of B7-H1 leads to inhibited T-cell response to and killing of tumor cells. Haematologica. 2013;98(9):1458–1466.
  • Harrington BK, Wheeler E, Hornbuckle K, et al. Modulation of immune checkpoint molecule expression in mantle cell lymphoma. Leuk Lymphoma. 2019;60(10):2498–2507.
  • Xerri L, Chetaille B, Seriari N, et al. Programmed death 1 is a marker of angioimmunoblastic T-cell lymphoma and B-cell small lymphocytic lymphoma/chronic lymphocytic leukemia. Hum Pathol. 2008;39(7):1050–1058.
  • Menter T, Bodmer-Haecki A, Dirnhofer S, et al. Evaluation of the diagnostic and prognostic value of PDL1 expression in Hodgkin and B-cell lymphomas. Hum Pathol. 2016;54:17–24.
  • Ek S, Björck E, Högerkorp CM, et al. Mantle cell lymphomas acquire increased expression of CCL4, CCL5 and 4-1BB-L implicated in cell survival. Int J Cancer. 2006;118(8):2092–2097.
  • Chao MP, Alizadeh AA, Tang C, et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell. 2010;142(5):699–713.
  • Chao MP, Tang C, Pachynski RK, et al. Extranodal dissemination of non-Hodgkin lymphoma requires CD47 and is inhibited by anti-CD47 antibody therapy. Blood Am Soc Hematol. 2011;118(18):4890–4901.
  • Jm R, A N, P H, et al. Infiltration of CD163-, PD-L1- and FoxP3-positive cells adversely affects outcome in patients with mantle cell lymphoma independent of established risk factors. Br J Haematol. 2021;193(3):520–531.
  • Aprile von Hohenstaufen K, Conconi A, de Campos CP, et al. Prognostic impact of monocyte count at presentation in mantle cell lymphoma. Br J Haematol. 2013;162(4):465–473.
  • Koh YW, Shin SJ, Park C, et al. Absolute monocyte count predicts overall survival in mantle cell lymphomas: correlation with tumour-associated macrophages. Hematol Oncol. 2014;32(4):178–186.
  • Nygren L, Wasik AM, Baumgartner-Wennerholm S, et al. T-Cell levels are prognostic in mantle cell lymphoma. Clin Cancer Res. 2014;20(23):6096–6104.
  • Immune-Depleted Tumor Microenvironment Signature Is Associated with BTK Inhibitor Resistance in Mantle Cell Lymphoma. American Society of Hematology; 2021.
  • Bernard S, Danglade D, Gardano L, et al. Inhibitors of BCR signalling interrupt the survival signal mediated by the micro-environment in mantle cell lymphoma. Int J Cancer. 2015;136(12):2761–2774.
  • Glofitamab Step-up Dosing Induces High Response Rates in Patients (pts) with Relapsed or Refractory (R/R) Mantle Cell Lymphoma (MCL), Most of Whom Had Failed Prior Bruton’s Tyrosine Kinase Inhibitor (BTKi) Therapy. ASH; 2021.
  • Wang M, Munoz J, Goy A, et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2020;382(14):1331–1342.
  • Morschhauser FA, Cartron G, Thieblemont C, et al. Obinutuzumab (GA101) monotherapy in relapsed/refractory diffuse large b-cell lymphoma or mantle-cell lymphoma: results from the phase II GAUGUIN study. J Clin Oncol. 2013;31(23):2912–2919.
  • Hagner PR, Chiu H, Ortiz M, et al. Activity of lenalidomide in mantle cell lymphoma can be explained by NK cell-mediated cytotoxicity. Br J Haematol. 2017;179(3):399–409.
  • Marriott J, Clarke I, Dredge K, et al. Thalidomide and its analogues have distinct and opposing effects on TNF-alpha and TNFR2 during co-stimulation of both CD4(+) and CD8(+) T cells. Clin Exp Immunol. 2002;130(1):75–84.
  • CD47-Blocker TTI-622 Shows Single-Agent Activity in Patients with Advanced Relapsed or Refractory Lymphoma: Update from the Ongoing First-in-Human Dose Escalation Study. Atlanta, Georgia: ASH; 2021.
  • Romaguera JE, Fayad LE, Feng L, et al. Ten-year follow-up after intense chemoimmunotherapy with Rituximab-HyperCVAD alternating with Rituximab-high dose methotrexate/cytarabine (R-MA) and without stem cell transplantation in patients with untreated aggressive mantle cell lymphoma. Br J Haematol. 2010;150(2):200–208.
  • Else M, Marín‐Niebla A, de la Cruz F, et al. Rituximab, used alone or in combination, is superior to other treatment modalities in splenic marginal zone lymphoma. Br J Haematol. 2012;159(3):322–328.
  • Torka P, Barth M, Ferdman R, et al. Mechanisms of resistance to monoclonal antibodies (mAbs) in lymphoid malignancies. Curr Hematol Malig Rep. 2019;14(5):426–438.
  • Hiraga J, Tomita A, Sugimoto T, et al. Down-regulation of CD20 expression in B-cell lymphoma cells after treatment with rituximab-containing combination chemotherapies: its prevalence and clinical significance. Blood J Am Soc Hematol. 2009;113(20):4885–4893.
  • Kuroda Y, Yashima-Abo A, Koyama D, et al. Bone marrow stromal cell-mediated degradation of CD20 leads to primary rituximab resistance in mantle cell lymphoma. Leukemia. 2021;35(5):1506–1510.
  • Dahal LN, Huang C-Y, Stopforth RJ, et al. Shaving is an epiphenomenon of type I and II anti-CD20-mediated phagocytosis, whereas antigenic modulation limits Type I monoclonal antibody efficacy. J Immunol. 2018;201(4):1211–1221.
  • Skopelja-Gardner S, Jones JD, Hamilton BJ, et al. Role for ZAP-70 signaling in the differential effector functions of rituximab and obinutuzumab (GA101) in chronic lymphocytic leukemia B cells. J Immunol. 2017;199(4):1275–1282.
  • Terui Y, Mishima Y, Sugimura N, et al. Identification of CD20 C-terminal deletion mutations associated with loss of CD20 expression in non-Hodgkin's lymphoma. Clin Cancer Res. 2009;15(7):2523–2530.
  • Golay J, Lazzari M, Facchinetti V, et al. CD20 levels determine the in vitro susceptibility to rituximab and complement of B-cell chronic lymphocytic leukemia: further regulation by CD55 and CD59. Blood. 2001;98(12):3383–3389.
  • Racila E, Link BK, Weng WK, et al. A polymorphism in the complement component C1qA correlates with prolonged response following rituximab therapy of follicular lymphoma. Clin Cancer Res. 2008;14(20):6697–6703.
  • Olejniczak SH, Hernandez-Ilizaliturri FJ, Clements JL, et al. Acquired resistance to rituximab is associated with chemotherapy resistance resulting from decreased Bax and Bak Expression. Clin Cancer Res. 2008;14(5):1550–1560.
  • Gu JJ, Singh A, Xue K, et al. Up-regulation of hexokinase II contributes to rituximab-chemotherapy resistance and is a clinically relevant target for therapeutic development. Oncotarget. 2018;9(3):4020–4033.
  • Heinrich DA, Weinkauf M, Hutter G, et al. Differential regulation patterns of the anti-CD20 antibodies obinutuzumab and rituximab in mantle cell lymphoma. Br J Haematol. 2015;168(4):606–610.
  • Wang ML, Lee H, Chuang H, et al. Ibrutinib in combination with rituximab in relapsed or refractory mantle cell lymphoma: a single-centre, open-label, phase 2 trial. Lancet Oncol. 2016;17(1):48–56.
  • Edelmann J, Dokal AD, Vilventhraraja E, et al. Rituximab and obinutuzumab differentially hijack the B cell receptor and NOTCH1 signaling pathways. Iscience. 2021;24(2):102089.
  • Habermann TM, Lossos IS, Justice G, et al. Lenalidomide oral monotherapy produces a high response rate in patients with relapsed or refractory mantle cell lymphoma. Br J Haematol. 2009;145(3):344–349.
  • Ruan J, Martin P, Shah B, et al. Lenalidomide plus rituximab as initial treatment for mantle-cell lymphoma. N Engl J Med. 2015;373(19):1835–1844.
  • Jerkeman M, Eskelund CW, Hutchings M, et al. Ibrutinib, lenalidomide, and rituximab in relapsed or refractory mantle cell lymphoma (PHILEMON): a multicentre, open-label, single-arm, phase 2 trial. Lancet Haematol. 2018;5(3):e109–e116.
  • Davies FE, Raje N, Hideshima T, et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood J Am Soc Hematol. 2001;98(1):210–216.
  • Reddy N, Hernandez‐Ilizaliturri FJ, Deeb G, et al. Immunomodulatory drugs stimulate natural killer-cell function, alter cytokine production by dendritic cells, and inhibit angiogenesis enhancing the anti-tumour activity of rituximab in vivo. Br J Haematol. 2008;140(1):36–45.
  • Chiu H, Trisal P, Bjorklund C, et al. Combination lenalidomide-rituximab immunotherapy activates anti-tumour immunity and induces tumour cell death by complementary mechanisms of action in follicular lymphoma. Br J Haematol. 2019;185(2):240–253.
  • Lagrue K, Carisey A, Morgan DJ, et al. Lenalidomide augments actin remodeling and lowers NK-cell activation thresholds. Blood J Am Soc Hematol. 2015;126(1):50–60.
  • Lopez-Girona A, Mendy D, Ito T, et al. Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide. Leukemia. 2012;26(11):2326–2335.
  • Galustian C, Meyer B, Labarthe MC, et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol Immunother. 2009;58(7):1033–1045.
  • Gandhi AK, Kang J, Havens CG, et al. Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN). Br J Haematol. 2014;164(6):811–821.
  • Ménard C, Rossille D, Dulong J, et al. Lenalidomide triggers T-cell effector functions in vivo in patients with follicular lymphoma. Blood Adv. 2021;5(8):2063–2074.
  • Menter T, Tzankov A, Zucca E, et al. Prognostic implications of the microenvironment for follicular lymphoma under immunomodulation therapy. Br J Haematol. 2020;189(4):707–717.
  • Zheng W, Carol E, Alli R, et al. PI3K orchestration of the in vivo persistence of chimeric antigen receptor-modified T cells. Leukemia. 2018;32(5):1157–1167.
  • Ruella M, Kenderian SS, Shestova O, et al. The addition of the BTK inhibitor ibrutinib to anti-CD19 chimeric antigen receptor T cells (CART19) improves responses against mantle cell lymphoma. Clin Cancer Res. 2016;22(11):2684–2696.
  • Rule S, Dreyling M, Goy A, et al. Ibrutinib for the treatment of relapsed/refractory mantle cell lymphoma: extended 3.5-year follow up from a pooled analysis. Haematologica. 2019;104(5):e211–e214.
  • Gauthier J, Hirayama AV, Purushe J, et al. Feasibility and efficacy of CD19-targeted CAR T cells with concurrent ibrutinib for CLL after ibrutinib failure. Blood. 2020;135(19):1650–1660.
  • Patel V, Balakrishnan K, Bibikova E, et al. Comparison of acalabrutinib, a selective bruton tyrosine kinase inhibitor, with ibrutinib in chronic lymphocytic leukemia cells. Clin Cancer Res. 2017;23(14):3734–3743.
  • Thijs WHF, Charnelle CT, Nan H, et al. Differential effects of BTK inhibitors ibrutinib and zanubrutinib on NK-cell effector function in patients with mantle cell lymphoma. Haematologica. 2020;105(2):e76–e79.
  • Phase 1/2 Trial of IL7/IL15-Expanded Bispecific LV20.19 CAR T-Cells for Relapsed, Refractory B-Cell Non-Hodgkin Lymphoma. Atlanta, Georgia: ASH; 2021.
  • Shah NN, Johnson BD, Schneider D, et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose escalation and expansion trial. Nat Med. 2020;26(10):1569–1575.
  • Wudhikarn K, Brunstein CG, Bachanova V, et al. Relapse of lymphoma after allogeneic hematopoietic cell transplantation: management strategies and outcome. Biol Blood Marrow Transplant. 2011;17(10):1497–1504.
  • Russell N, Byrne J, Faulkner R, et al. Donor lymphocyte infusions can result in sustained remissions in patients with residual or relapsed lymphoid malignancy following allogeneic haemopoietic stem cell transplantation. Bone Marrow Transplant. 2005;36(5):437–441.
  • Bloor AJ, Thomson K, Chowdhry N, et al. High response rate to donor lymphocyte infusion after allogeneic stem cell transplantation for indolent non-Hodgkin lymphoma. Biol Blood Marrow Transplant. 2008;14(1):50–58.
  • Tam CS, Bassett R, Ledesma C, et al. Mature results of the MD anderson cancer center risk-adapted transplantation strategy in mantle cell lymphoma. Blood J Am Soc Hematol. 2009;113(18):4144–4152.
  • Urbano-Ispizua A, Pavletic SZ, Flowers ME, et al. The impact of graft-versus-host disease on the relapse rate in patients with lymphoma depends on the histological subtype and the intensity of the conditioning regimen. Biol Blood Marrow Transplant. 2015;21(10):1746–1753.
  • Cook G, Smith GM, Kirkland K, et al. Outcome following Reduced-Intensity Allogeneic Stem Cell Transplantation (RIC AlloSCT) for relapsed and refractory mantle cell lymphoma (MCL): a study of the British Society for blood and marrow transplantation. Biol Blood Marrow Transplant. 2010;16(10):1419–1427.
  • Robinson SP, Boumendil A, Finel H, et al. Long-term outcome analysis of reduced-intensity allogeneic stem cell transplantation in patients with mantle cell lymphoma: a retrospective study from the EBMT lymphoma working party. Bone Marrow Transplant. 2018;53(5):617–624.
  • Tessoulin B, Ceballos P, Chevallier P, et al. Allogeneic stem cell transplantation for patients with mantle cell lymphoma who failed autologous stem cell transplantation: a national survey of the SFGM-TC. Bone Marrow Transplant. 2016;51(9):1184–1190.
  • Lin RJ, Ho C, Hilden PD, et al. Allogeneic haematopoietic cell transplantation impacts on outcomes of mantle cell lymphoma with TP 53 alterations. Br J Haematol. 2019;184(6):1006–1010.
  • Hutchings M, Morschhauser F, Iacoboni G, et al. Glofitamab, a novel, bivalent CD20-Targeting T-Cell-Engaging bispecific antibody, induces durable complete remissions in relapsed or refractory B-Cell lymphoma: a phase I trial. J Clin Oncol. 2021;39(18):1959–JCO2003175.
  • Hutchings M, Mous R, Clausen MR, et al. Dose escalation of subcutaneous epcoritamab in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: an open-label, phase 1/2 study. Lancet. 2021;398(10306):1157–1113.
  • Budde LE, Sehn LH, Assouline S, et al. Mosunetuzumab, a Full-Length bispecific CD20/CD3 antibody, displays clinical activity in relapsed/refractory B-Cell Non-Hodgkin lymphoma (NHL): interim safety and efficacy results from a phase 1 study. Blood. 2018;132(Supplement 1):399–399.
  • Bannerji R, Allan JN, Arnason JE, et al. Odronextamab (REGN1979), a human CD20 x CD3 bispecific antibody, induces durable, complete responses in patients with highly refractory B-Cell Non-Hodgkin lymphoma, including patients refractory to CAR T therapy. Blood. 2020;136(Supplement 1):42–43.
  • van der Horst HJ, de Jonge AV, Hiemstra IH, et al. Epcoritamab induces potent anti-tumor activity against malignant B-cells from patients with DLBCL, FL and MCL, irrespective of prior CD20 monoclonal antibody treatment. Blood Cancer J. 2021;11(2):38.
  • Jurczak W, Zinzani PL, Hess G, et al. A phase IIa, open-label, multicenter study of single-agent tafasitamab (MOR208), an Fc-optimized anti-CD19 antibody, in patients with relapsed or refractory B-cell non-Hodgkin's lymphoma: long-term follow-up, final analysis. Washington, DC: American Society of Hematology; 2019.
  • Lee HJ, Choi MY, Siddiqi T, et al. Phase 1/2 study of cirmtuzumab and ibrutinib in mantle cell lymphoma (MCL) or chronic lymphocytic leukemia (CLL). J Clin Oncol. 2021;39(15_suppl):7556–7556.
  • Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase Ib study. J Clin Oncol. 2016;34(23):2698–2704.
  • Ansell SM, Hurvitz SA, Koenig PA, et al. Phase I study of ipilimumab, an anti-CTLA-4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma. Clin Cancer Res. 2009;15(20):6446–6453.
  • Frank MJ, Khodadoust MS, Czerwinski DK, et al. Autologous tumor cell vaccine induces antitumor T cell immune responses in patients with mantle cell lymphoma: a phase I/II trial. J Exp Med. 2020;217(9):e20191712.
  • Kim TM, Lakhani N, Gainor J, et al. ALX148, a CD47 blocker, in combination with rituximab in patients with Non-Hodgkin lymphoma. Blood. 2020;136(Supplement 1):13–14.
  • Advani R, Bartlett NL, Smith SM, et al. The first-in-class anti-cd47 antibody hu5f9-G4 + rituximab induces durable responses in relapsed/refractory DLBCL and indolent lymphoma: interim phase 1B/2 results. Hematol Oncol. 2019;37:89–90.
  • Lokhande L, Emruli VK, Kolstad A, et al. Immune-related protein signature in serum stratify relapsed mantle cell lymphoma patients based on risk. BMC Cancer. 2020;20(1):1–13.
  • Guo A, Lu P, Coffey G, et al. Dual SYK/JAK inhibition overcomes ibrutinib resistance in chronic lymphocytic leukemia: cerdulatinib, but not ibrutinib, induces apoptosis of tumor cells protected by the microenvironment. Oncotarget. 2017;8(8):12953–12967.
  • Blunt MD, Koehrer S, Dobson RC, et al. The dual syk/JAK inhibitor cerdulatinib antagonizes B-cell receptor and microenvironmental signaling in chronic lymphocytic leukemia. Clin Cancer Res. 2017;23(9):2313–2324.
  • Papin A, Tessoulin B, Bellanger C, et al. CSF1R and BTK inhibitions as novel strategies to disrupt the dialog between mantle cell lymphoma and macrophages. Leukemia. 2019;33(10):2442–2453.
  • Zhao X, Lwin T, Silva A, et al. Unification of de novo and acquired ibrutinib resistance in mantle cell lymphoma. Nat Commun. 2017;8(1):14920–14915.
  • Guan J, Huang D, Yakimchuk K, et al. p110α inhibition overcomes stromal cell-mediated ibrutinib resistance in mantle cell lymphoma. Mol Cancer Ther. 2018;17(5):1090–1100.
  • Eyre TA, Walter HS, Iyengar S, et al. Efficacy of venetoclax monotherapy in patients with relapsed, refractory mantle cell lymphoma after Bruton tyrosine kinase inhibitor therapy. Haematologica. 2019;104(2):e68–e71.
  • Jayappa KD, Portell CA, Gordon VL, et al. Microenvironmental agonists generate de novo phenotypic resistance to combined ibrutinib plus venetoclax in CLL and MCL. Blood Adv. 2017;1(14):933–946.
  • Kaplan LD, Maurer MJ, Stock W, et al. Bortezomib consolidation or maintenance following immunochemotherapy and autologous stem cell transplantation for mantle cell lymphoma: CALGB/alliance 50403. Am J Hematol. 2020;95(6):583–593.
  • Till BG, Li H, Bernstein SH, et al. Phase II trial of R-CHOP plus bortezomib induction therapy followed by bortezomib maintenance for newly diagnosed mantle cell lymphoma: SWOG S0601. Br J Haematol. 2016;172(2):208–218.
  • Oppermann S, Ylanko J, Shi Y, et al. High-content screening identifies kinase inhibitors that overcome venetoclax resistance in activated CLL cells. Blood J Am Soc Hematol. 2016;128(7):934–947.
  • Haselager M, Thijssen R, West C, et al. Regulation of Bcl-XL by non-canonical NF-κB in the context of CD40-induced drug resistance in CLL. Cell Death Differ. 2021;28(5):1658–1668.

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.