Supplementary Material and Methods Cells, transfection and transduction MOPC315 was obtained as an in vitro-adapted cell line from ATCC (Manassa, VA). The Id non-secreting variant MOPC315.36 was provided by Alexander Marks, University of Toronto. The corresponding bone-marrow homing MOPC315.BM cell line was obtained by serial in vivo passage with selection of tumor cells growing within the bone marrow, as previously described.1 The MOPC315.BM-luc2P variant, expressing the Photinus pyralis firefly luciferase, were generated by lentiviral transduction with the pHIV-LucPZsGreen vector, provided by Dr. Bryan Welm, University of Utah (Provided through the Addgene repository, Addgene cat. #39196). Lentiviral particle were generated by transient transfection of HEK293T cells with Lipofectamine2000 (Life Technologies) with the relevant expression vector together with the packaging vector pCMV-VSV-g and pCMV-dR8.2dvpr (Addgene #8454 and #8455; generously provided by Dr. Robert Weinberg, Whitehead Institute). Id-specific T cells were isolated by negative selection by magnetic beads using the CD4+ T Cell Isolation Kit II (Miltenyi Biotech GmbH) according to the supplied protocol. In vitro activation, Th1 polarization and expansion of Id- and DO11.10-specific CD4+ T cells was performed as previously described.2, 3 Analysis of cells by flow cytometry Single-cell suspensions from femurs were obtained by use of a stainless steel sieve. Unspecific binding was blocked by incubation with heat-inactivated (56 °C, 30 min) 30% normal rat serum in PBS and 100 μg ml−1 anti-FcγRII/III monoclonal antibodies (mAb; clone 2.4G2). Cells were stained for 15 min on ice with specific mAbs in PBS supplemented with 0.5% bovine serum albumin (Biotest). The following commercially available mAbs were used, conjugated with either FITC, PE, or Pe-Cy7: CD3 (17A2, 561798), CD4 (GK1.5, 563933), IFN-γ (XMG1.2, 562020), IL-4 (11B11, 560699), IL-17 (TC11-18H10, 559502), MHC class II I-E (14-4-4S, 553544) (BD Biosciences), Gata-3 (TWAJ, 12-9966-41), T-bet (4B10, 12-5825), and RORgt (B2D, 12-6981-80) (eBioscience). The following mAbs were affinity-purified and, if needed, biotinylated in our laboratory: anti-Id-specific-TCR-clonotype (GB113), anti-FcγRII/III (2.4G2; ATCC). Biotinylated mAbs were detected with streptavidin conjugated to peridinin chlorophyll protein (BD Biosciences). For intracellular cytokine detection, cells were stimulated with phorbol myristate acetate and ionomycin (both from Sigma) in vitro for 4 h in cell culture medium supplemented with monensin, before staining with Cytofix/Cytoperm Plus reagents (BD Biosciences) and specific mAbs. Cells were analyzed on a Fortessa instrument with FACSDiva software (BD Biosciences). Mice Id-specific TCR-transgenic mice on BALB/c and BALB/c Rag1-/- backgrounds have been previously described.4, 5 BALB/c and BALB/c Rag1-/- mice were obtained from Taconic (Taconic Biosciences Inc., Ry, Denmark). BALB/c nu/nu mice were purchased from Janvier Labs (Saint-Berthevin, France). All experiments were performed with equal amounts of male and female mice aged 8-12 weeks at experiment start. Experiments were performed according to institutional and governmental guidelines and were approved by the National Committee for Animal Experiments (Oslo, Norway). MHC II I-Ed knockout by CRISPR/Cas9 MOPC315.BM cells deficient in MHC II expression was achieved by introduction of a non-sense mutation within the I-Ed beta chain using a CRISPR/Cas9 approach. Two sgRNA sequences (5′-GCCACACCATGCTGCAGGAG-3′, 5'-GACAGTGCTGAGCCCTCCAG3') were designed to target sites within the H2-eb1 gene (Gene ID: 14969) and inserted into the CRISPR/Cas9 expression vector PX458 (a kind gift from Feng Zhang, Broad Institute; Addgene plasmid #48138). Cells were transiently transfected with PX458 using an Amaxa Nucleofector (Lonza Inc., Basel, Switzerland). GFP-expressing clones were isolated by fluorescence-activated cell sorting using a FACS Aria II (BD Bioscience). Clones were screened for mutations by PCR amplification and target sequencing, and clones harboring non-sense mutations in both alleles were selected. Targeting sequence functionality was verified by transfection of an MHC II-positive B lymphoma cell line. Tumor challenge, in vivo imaging and adoptive T cell transfer Tumor cells were washed, suspended in PBS, and the indicated numbers of cells injected i.v. in the lateral tail vein. Mice were observed twice weekly for disease, and long-term monitored mice euthanized upon development of paraplegia, or weight loss > 15%. Tumor burden was quantified by measurement of serum M315 myeloma protein by a sandwich ELISA as previously described.3 In vivo bioluminescence imaging was performed under isoflurane anaesthesia using an IVIS Spectrum imaging system (Xenogen Corp), as previously described.1 The threshold of bioluminescence signals was automatically determined using the Living Image software package (Xenogen Corp.). Bioluminescence was quantified as photons/sec/cm2/steradian after subtraction of the background luminescence from control mice not injected with tumor cells, but that had received the luciferin substrate. For monitoring of firefly luciferase activity, D-luciferin (Biosynth AG, Staad, Switzerland) was injected i.p. (150 mg/kg body weight). Adoptive transfer of T cells was performed by i.v. injection of the indicated number of cells suspended in 100L PBS. For indicated experiments, wild-type BALB/c mice were lymphodepleted (500cGy) using an Xstrahl RS320 X-Ray irradiator (Xstrahl Ltd, Surrey, England) 24h prior to T cell transfer. Immunohistochemistry Specimens for histochemistry and immunofluorescence staining were fixed for 24 hours in 4% buffered parafomaldehyde. Bones were further processed in decalcifying buffer (0.38 M EDTA, 0.36 M NaOH pH 7.0) before paraffin embedding. 4m sections were prepared, de-waxed with xylene, and rehydrated. Antigen retrieval was performed by 1h incubation in Dako buffer (pH10.0; Dako, Glostrup, Denmark) or buffered sodium citrate (pH 6.0) in a 100C water bath. Sections were analyzed by microscopy (Nikon Eclipse E 800) and digital images acquired and processed with AnalySIS Soft Imaging System. Statistical analysis For tumor challenge experiments, differences in survival were analysed with the logrank test. Total flux data were analyzed with the Mann–Whitney test. Statistical analysis was performed with GraphPad Prism 5 software. References 1. Hofgaard PO, Jodal HC, Bommert K, Huard B, Caers J, Carlsen H, et al. A Novel Mouse Model for Multiple Myeloma (MOPC315.BM) That Allows Noninvasive Spatiotemporal Detection of Osteolytic Disease. PloS one 2012; 7(12): e51892. 2. Haabeth OAW, Lorvik KB, Hammarström C, Donaldson IM, Haraldsen G, Bogen B, et al. Inflammation driven by tumour-specific Th1 cells protects against B-cell cancer. Nature Communications 2011 Apr 15; 2: 240. 3. Lauritzsen GF, Weiss S, Dembic Z, Bogen B. Naive idiotype-specific CD4+ T cells and immunosurveillance of B-cell tumors. Proceedings of the National Academy of Sciences of the United States of America 1994 Jul 07; 91(12): 5700-5704. 4. Bogen B, Gleditsch L, Weiss S, Dembic Z. Weak positive selection of transgenic T cell receptor-bearing thymocytes: importance of major histocompatibility complex class II, T cell receptor and CD4 surface molecule densities. European journal of immunology 1992 Apr; 22(3): 703-709. 5. Lundin KU, Screpanti V, Omholt H, Hofgaard PO, Yagita H, Grandien A, et al. CD4+ T cells kill Id+ B-lymphoma cells: FasLigand-Fas interaction is dominant in vitro but is redundant in vivo. Cancer immunology, immunotherapy : CII 2004 Dec 01; 53(12): 11351145.