Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Apr 2.
Published in final edited form as: Breast Cancer Res Treat. 2022 Nov 1;197(1):57–69. doi: 10.1007/s10549-022-06783-1

EGFR as a potent CAR T target in triple negative breast cancer brain metastases

Siddharth Subham 1,2,3, John D Jeppson 1, Colette Worcester 1,4, Bryan Schatmeyer 5, Jie Zhao 2, Rashna Madan 4, Nelli S Lakis 4, Bruce F Kimler 1, Joseph P McGuirk 6, Ronald C Chen 1, Shane R Stecklein 1,2,4, David Akhavan 1,2,3
PMCID: PMC10987173  NIHMSID: NIHMS1972641  PMID: 36318382

Abstract

Purpose

There is currently no curative treatment for patients diagnosed with triple-negative breast cancer brain metastases (TNBC-BM). CAR T cells hold potential for curative treatment given they retain the cytolytic activity of a T cell combined with the specificity of an antibody. In this proposal we evaluated the potential of EGFR re-directed CAR T cells as a therapeutic treatment against TNBC cells in vitro and in vivo.

Methods

We leveraged a TNBC-BM tissue microarray and a large panel of TNBC cell lines and identified elevated epidermal growth factor receptor (EGFR) expression. Next, we designed a second-generation anti-EGFR CAR T construct incorporating a clinically relevant mAb806 tumor specific single-chain variable fragment (scFv) and intracellular 4–1BB costimulatory domain and CD3ζ using a lentivirus system and evaluated in vitro and in vivo anti-tumor activity.

Results

We demonstrate EGFR is enriched in TNBC-BM patient tissue after neurosurgical resection, with six of 13 brain metastases demonstrating both membranous and cytoplasmic EGFR. Eleven of 13 TNBC cell lines have EGFR surface expression ≥ 85% by flow cytometry. EGFR806 CAR T treated mice effectively eradicated TNBC-BM and enhanced mouse survival (log rank p < 0.004).

Conclusion

Our results demonstrates anti-tumor activity of EGFR806 CAR T cells against TNBC cells in vitro and in vivo. Given EGFR806 CAR T cells are currently undergoing clinical trials in primary brain tumor patients without obvious toxicity, our results are immediately actionable against the TNBC-BM patient population.

Keywords: Breast cancer, TNBC, Brain metastases, CAR T, EGFR

Introduction

Breast cancer is the most frequently diagnosed cancer among American females and is the second leading cause of cancer-related deaths with a predicted 43,250 deaths this year [1]. Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer which is defined by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. It accounts for 15–20% of all breast cancer cases and is known for its high recurrence rate and high ability to metastasize resulting in poor clinical prognosis [2]. The most common systemic therapy for metastatic TNBC (mTNBC) is chemotherapy [3], but median overall survival (OS) is limited to 12 to 18 months [4]. Only 20–40% of mTNBC are PD-L1 positive and candidates for checkpoint inhibitor immunotherapy [5, 6]. Worse yet, the incidence of brain metastases (TNBC-BM) is as high as 46% among patients with advanced TNBC[7]. Median survival from the diagnosis of TNBC-BM is only 4.9 months [7, 8]. Despite many clinical trials, no current standard therapy exists that provides long-term control or cure for TNBC-BM patients [7, 9, 10].

Chimeric Antigen Receptor(CAR) T cell therapy combines the cytolytic potency of a T cell with the antigen specificity of an antibody single-chain (scFv) domain. Although CAR T cells have had remarkable clinical success in liquid tumors [11, 12], significant hurdles still exist in their application against brain tumors[13], a major obstacle being identifying appropriate tumor antigen that has low to no expression in normal tissues. EGFR is highly expressed in 13–76% [1419] of TNBC. Monoclonal antibody [2022] and small-molecule targeted therapy against EGFR [23, 24] have failed in mTNBC, due to the lack of cytolytic-effect, the activation of downstream compensatory oncogenic signaling pathways, and the co-expression of other onco-receptors [2527]. CAR T cells targeting breast-to-brain metastases hold promise due to their cytolytic effect against tumor cells. Preclinical models of HER2 CAR T cells have shown anti-tumor activity in HER2 + xenograft mouse models of breast-to-brain metastases [28]. As a result, HER2 + breast-to-brain metastatic patients are undergoing clinical study for efficacy of intracranial delivered HER2 + CAR T cells (NCT03696030). However, there are currently no clinical trials investigating CAR T cells targeting TNBC-BM patients. EGFR may be an attractive target in TNBC-BM tissue. It is known that TNBCs expressing EGFR are prone to metastasize to the brain compared to those lacking EGFR [29]. In a study of 33 breast cancer patients, EGFR expression was found in 39% patients who had brain metastasis [30], although the percentage of patients with TNBC was not stated. Herein, we explore the percentage expression of EGFR in primary TNBC-BM tissue, as well as evaluate the preclinical efficacy of EGFR as a CAR T target in TNBC-BM mouse models.

EGFR CAR T cells have been explored in preclinical TNBC models [31, 32], however, the scFv is based on the anti-EGFR cetuximab antibody that targets both oncogenic EGFR and skin-keratinocyte EGFR [33, 34], thus raising concern for on-target off-tumor toxicity. Indeed, studies in cetuximab based EGFR CAR T demonstrate toxicity in human skin orthotopic xenografts [35]. To address this problem, we employed a CAR T construct employing the EGFR mAb806 antibody (EGFR806), which is tumor-restricted to EGFR as a result of oncogene amplification [3638]. Furthermore, mAb806 kills TNBC cells in vitro when conjugated with toxin [39]. The mAb806 antibody has been tested in clinical trials in EGFR + tumors with mild toxicity [40]. Intracranial delivered EGFR806 CAR T cells are currently under clinical study in pediatric brain tumors (NCT03638167), with no dose limiting toxicity in preliminary results [41]. Further, EGFR expression in normal adult human brain is low to negligible, reducing the risk of on-target off-tumor toxicity [4244].

Here, we have developed second generation EGFR806 CAR T cells for the treatment of TNBC-BM. To our knowledge, this is the first CAR T cell targeting metastatic TNBC using this mAb806 derived scFv. We evaluated therapeutic efficacy of intracranial tumoral (ICT) delivery of CAR T cells at the site of tumor in orthotopic human tumor xenograft models. Our data provide support for potential clinical application of EGFR806 CAR T cells targeting TNBC-BM in human clinical trials.

Materials and methods

Cell culture

Human TNBC cell lines MDA-MB-231, MDA-MB-468, MDA-MB-436, MDA-MB-453, HCC70, HCC1937, HCC1143, HCC1187, HCC1395, HS578T, BT549 and BT20 were a gift from Dr. Shane Stecklein (collaborator). MDA-MB-231 and MDA-MB-468 were maintained in DMEM media (10–013-CM; Corning, Inc., Corning, NY) supplemented with 10% Fetal Bovine Serum (FBS) (900–108, Gemini BioProducts; W. Sacramento, CA) and 1% Pen-Strep (15070*063; Gibco; Thermo Fisher Scientific, Waltham, MA). HEK-293T cells (LV-Max; Takara Bio, San Jose, CA) were cultured in the same media and supplementation as TNBC cells. HCC70 cells were maintained in RPMI media (10–040-CM; Corning) with the same supplements as previous. All cells were maintained in a 37 °C incubator at 5% CO2 and were used within 20 passages. All cell lines were mycoplasma tested (Plasmotest, Invivogen; San Diego CA). All cell lines were counted using a Guava Muse® Cell Analyzer (Luminex Corporation, Houston, TX), with Count and Viability kit (MCH600103, Luminex Corp).

EGFR806 CAR T construct design

The EGFR806 targeted scFv sequence was derived from antibody Mab806 and cloned into second generation CAR T construct (Fig. 2A). A second generation construct was employed, as this format has demonstrated safety in multiple brain tumor related clinical trials. The extracellular spacer domain comprised of the double mutated IgG4EQ [45], followed by CD4 transmembrane domain, 41BB co-stimulatory domain and CD3ζ cytolytic domain. The CAR construct was cloned in a pLenti6.3/V-5-DEST backbone under the control of CMV promoter (GeneArt;Thermo Fisher Scientific, Waltham, MA). Full length amino acid sequence is described in supplemental Table 2.

Fig. 2.

Fig. 2

EGFR806 CAR T production for in vitro and in vivo evaluation. a. Second generation EGFR806 CAR design containing IgG4EQ hinge, CD4 Transmembrane domain, 4–1BB costimuatory domain followed by CD3zeta. b. Human PBMC derived CD4 + lentivirus transduced CAR T cells were thawed day 8, confirmed cell viability and CAR expression, and implanted ICT. c. Gating strategy for confirmation of CAR T cell expression: T cell population selected excluding debris in FSC v SSC, single cells were then selected by comparing FSC area v height and then live cells by DAPI. CARs were stained using anti-IgG (Jackson) and the gate was set using mock transduced T cells. d. Histogram demonstrates DAPI stain histogram of tumor only (gray), tumor + mock T cells (blue), tumor + EGFR806 CAR T (red). e. Gating strategy for MDA-MB-231 incubated 1:1 with mock T cells at 96 h. Tumor cells were gated to exclude debris and T cell populations. Single cells were then selected to prevent background positivity and the viability gate was set based on Tumor + Mock T cell population

Lentivirus production

HEK293T cells (Takara) were plated in T-300 flasks (TP90301; Midwest Scientific, St. Louis, MO), a day prior to transfection of the CAR and enhanced Green Fluorescent Protein-Firefly-Luciferase (eGFP-Ffluc, 119816; AddGene, Watertown, MA) plasmid constructs separately. Transfection was done at 70% confluence using CalPhos Mammalian Transfection Kit (631312; Takara Bio) with LV-MAX Lentiviral Packaging Mix (A43237; GibcoTM). Media was renewed the next day morning with addition of 0.5 mM Sodium butyrate ((B5887; Sigma-Aldrich, St. Louis, MO) and supernatant collected in 50 mL tubes 72 h later. The tubes were centrifuged at 800 g for 10 min to remove cellular debris and filtered through a 0.45 μm filter. The supernatants were combined and centrifuged at 6080 g at 4°C for 24 h. The lentivirus pellet was resuspended in 4% Lactose (L5–500; Fisher Chemical; Thermo Fisher Scientific, Waltham, MA, USA) in PBS (10,010–031; Gibco) solution, aliquoted and stored in −80°C freezer for further use. All chemical solutions were filtered in 0.2 μm.

Human CAR T cell production

Peripheral blood samples from healthy human male and female patients < 45 years of age without prior cancer diagnosis were collected in vacutainer tubes ± EDTA, followed by centrifugation at 4 °C set at 1300 g for 10 min, followed by collection of buffy coat. Untouched human CD4 T cells were isolated from buffy coat using Dynabeads® (11346D; Invitrogen; Thermo Fisher Scientific, Waltham, MA) according to the manufacturer’s instructions. Briefly, RBC lysis buffer was added to the buffy coat to lyse red blood cells and then magnetic beads were added to the PBMCs. After washing and spinning down tagged cells, Dynabeads® were added to the cells to negatively select the CD4 population. Finally, the supernatant was centrifuged to collect untouched CD4 T cells. The T cells were then cultured in gas-permeable tissue 24-well culture plates (80192 M; Wilsonwolf, St Paul, MN) at a concentration of 2 × 106 cells/well in 1 mL of LymphoONE T-Cell Expansion Xeno-Free media (WK552S; Takara) supplemented with 10% FBS, 50 U/mL IL2 (200–02; Peprotech, Cranbury, NJ), 0.5 ng/mL IL15 (200–15; peprotech) and one-time 20μL T Cell Trans-Act (130–128–758; Miltenyi Biotec, Gaithersburg, MD). EGFR806 CAR T lentivirus particles are added on D1 at multiplicity of infection (MOI) of 6. Media was doubled on D2 (2 × cytokines were added on this day only) & D3, and 3 mL added on D4 for a total of 7 mL. On D5 and D6 half of the media was replaced with fresh media (1 × cytokines were added in each of these days). Cells were collected on Day 7, followed by CAR expression by flow cytometry, then frozen down for later use in cryopreservation media (07930; CryoStor® StemCell Technologies, Cambridge, MA). CAR expression was identified by flow cytometry using anti-human IgG, Fcγ fragment specific (109–546–008 Jackson Immuno Research, West Grove, Pennsylvania).

Generation of stable TNBC cell lines expressing firefly luciferase

MDA-MB-231 cells were transduced with lentivirus to express eGFP-Ffluc under Puromycin (VWR) selection. Cells were then flow sorted (FACs Aria II, BD Biosciences, San Jose, CA) based on GFP expression (90%) and cultured as described above while maintaining puromycin selection.

Flow cytometry

For flow cytometry, live cells were detached using Cell-Stripper (Corning) and cells were then suspended in FACS Stain Solution (FSS) (PBS w/o CaCl2 & MgCl2, 0.1%BSA (15260–037; Gibco), 0.5 mM EDTA (351–027–721; Quality Biological, Gaithersburg, MD) and washed twice before and after adding anti-EGFR antibody (352907; Biolegend, San Diego CA) or isotype control. Cells were incubated with respective antibodies for 30 min in the dark at 4°C. Flow cytometry was run on BD LSRII (BD Biosciences) and cell viability was determined using 4′, 6-diamidino-2-phenylindole (DAPI, Sigma) (D9542; Sigma-Aldrich) or Ghost Dye Red 780 Viability Dye (13–0865-T100; Tonbo Biosciences; San Diego, CA). Data were then analyzed using FlowJo software (v10.7.1; FlowJo; BD Biosciences).

For Perforin and Granzyme CAR T activation assays, 50,000 MDA-MB-231 cells were plated per well of a 48 well plate and allowed to attach overnight. 100,000 CD4 CAR T cells and untransduced Mock T cells were added to the cells the following morning and allowed to incubate for 3 h. Brefeldin A was added for to the co-culture for another 5 h after which the T cells were collected in a V-Bottom 96 well plate and cells were washed thrice before adding extracellular antibodies and allowed to incubate at 4C for 30 min. After 2 washes, cells were fixed with Fixation Buffer (BioLegend #420801) in the dark for 20 min at room temperature. Cells were centrifuged at 350 g for 5 min and washed with Cell Staining Buffer. Samples were left with Cell Staining Buffer on them and left at 4C overnight. The fixed cells were suspended in Intracellular staining Perm Wash Buffer (BioLegend #421002) and centrifuged at 350 g for 5 min twice. The cells were than incubated with antibodies against Perforin (Biolegend #353312) and GranzymeB (Biolegend #372208) in Perm Wash Buffer for 20 min in the dark at room temperature. Cells were washed twice with Intracellular staining Perm Wash Buffer and centrifuged at 350 g for 5 min. Cells were finally resuspended in FSS and Flow was run.

Immunoblot

TNBC cell lines and CRISPR KO Lines were interrogated for EGFR expression by Immunoblot. In brief, cell lines were grown to confluence and lysed using RIPA buffer (89901; Thermo Fisher Scientific), with Protease and Phosphatase inhibitor (1861281; Thermo Fisher Scientific) and EDTA (1861274; Thermo Fisher Scientific). These lysates had protein concentrations quantified using a BCA Protein Assay Kit (23227; Thermo Fisher Scientific) on a Nanodrop (ND-2000; Thermo Fisher Scientific) according to manufacture instructions. Lysates were then prepared and run using Separation (SM-W001; Protein Simple, Minneapolis, MN) and Detection (DM-001 & DM-002; Protein Simple) Kits and manufacture instructions for the Protein Simple WES (004–600; Protein Simple) following the manufacturer’s instructions with primary antibodies for EGFR (4267T; Cell Signaling Technology, Danvers, MA) Actin (MAB8929; R&D Systems, Minneapolis, MN) and Vinculin (MAB6896-SP; Bio-Techne, Minneapolis, MN). Results were analyzed using Compass for SW software (Protein Simple).

In vitro killing assay

Human TNBC cells were co-cultured with EGFR806 CAR T cells and mock T cells at specified effector cell: TNBC cell ratios. After the specified duration of co-culture, the supernatant is collected in tubes and attached tumor cells detached using Cellstripper (25–056-CI; Corning) for 15–30 min incubation while shaking 37C. All cells are transferred to tubes and were stained for CD3, and a Live/Dead viability dye as described above for evaluating killing by the CAR T cells. CAR T cells were identified by anti-IgG to target hinge region of CAR (109–096–008; JacksonImmuno Research Labs). Flow cytometry was acquired on Cytek Aurora and data analyzed using FlowJo software.

Mouse tumor studies

Female NSG (NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ) mice, 8–11 weeks of age were purchased from the Jackson Laboratory (Jackson Laboratory; Farmington, CT). All mice experiments were approved by the Institutional Animal Care and Use Committee (IACUC).

The intracranial implantation of tumor cells has been previously described [46]. Briefly, mice were anesthetized by exposure to isoflurane. A burr hole was drilled on the skull at 2 mm lateral of the center line, 0.5 mm anterior to the bregma and 1 × 105 MDA-MB-231 cells expressing luciferase suspended in 3ul of PBS was injected orthotopically in 1 ul increments at 2.5 mm, 2 mm, and 1.5 mm deep from the dura. Engraftment was verified by bioluminescent imaging (IVIS Spectrum imager; PerkinElmer, Waltham, MA) using intraperitoneal injection of 200ul (100 mM) D-Luciferin one day before CAR T cell injection. The mice were randomized into groups based on the BLI signal and 1 × 106 EGFR806 CAR T cells or mock treatment injected intracranially at the same burr hole at 0.5ul per incremental depth (2.75 mm, 2.5 mm, 2.25 mm, 1.75 mm, 1.5 mm, & 1.25 mm) site next day. Tumor growth was monitored by IVIS imager and flux signals analyzed using Living Image®software (v4.5.5; PerkinElmer). Mice with BLI negative tumors underwent repeat BLI imaging to confirm absence of BLI signal. Additional dates for luminescent imaging were chosen to capture the exponential growth of tumor in non-treated mice based on previous work with the tumor kinetics. After the control mice were all euthanized, imaging was halted and mice were monitored for survival. At D100 the mice were imaged once more. At this time the luminescence seen in two CAR treated mice had disappeared leading to them to be imaged again in the subsequent timepoints after the machine had been serviced to ensure that it was still functioning. At desired time points or at moribund status, mice were euthanized, and brain tissues were processed for H&E histology as described below.

Immunohistochemistry staining

A human tissue microarray was constructed from resected brain metastases of 13 patients in duplicate and two positive controls (placenta, and kidney) and one negative control (tonsil) for EGFR expression. Pathologist confirmed tissue prior to TMA construction, and pathologist confirmed IHC scoring. Anti-EGFR antibody (Emab-134 ab264540 Abcam; Cambridge, UK) was used for IHC staining according to manufacturer instruction. EGFR immunoreactivity was scored by a clinical pathologist and quantified based on the percentage of tumor cells exhibiting weak (1 +), moderate (2 +), or strong (3 +) intensity staining of cell surface and cytoplasmic staining. The H score is obtained by the formula: (3 × percentage of strongly staining cells) + (2 × percentage of moderately staining cells) + percentage of weakly staining cells, giving a range of 0 to 300. Each patient was assigned a separate H-score for both membrane and cytoplasmic staining.

For murine H&E, mice were euthanized at indicated time points and were perfused with ice cold PBS followed by 4% PFA. Whole brains were dissected and incubated in 4% PFA for 3 days, followed by 70% ethanol for 3 days before being embedded in paraffin. Transverse Sects. (10 μm thick) were cut and stained with hematoxylin and eosin.

Statistical analysis

All statistical analysis was performed using Prism software (GraphPad v9). Data are represented as Mean ± SD as stated in the figure legends. Biological significance was determined by student’s T-test or using one-way ANOVA with multiple comparisons. For mouse survival studies, differences between groups were assessed by log-rank (Mantle-Cox) test. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.

Results

EGFR is highly expressed in TNBC-BM patient tissues and cell lines

We generated a tissue microarray of resected metastatic brain tumor tissue for 13 TNBC-BM patients and H-score was assigned individually for both surface and cytoplasmic EGFR expression (Fig. 1A, supplemental Fig s1 and Table 1). This result demonstrates high membranous and cytoplasmic expression of EGFR in 6 of 13 patients. Strikingly, 3 patients have near maximum membranous H-score of 300. Next, we evaluated surface expression of EGFR in a panel of TNBC breast cancer lines (Fig. 1B). Eleven of 13 tested cell lines had EGFR mean fluorescence intensity (MFI) greater than 85%, and only one cell line (MDA-MB-453) was negative for EGFR. In order to confirm these findings, we also interrogated the 13 cell lines by immunoblot. 12 of 13 cell lines demonstrated high levels of EGFR expression relative to EGFR knockout control line (Fig. 1C). We chose three high EGFR expressing cell lines (MDA-MB-231, MDA-MB-468 and HCC70) representing three different intrinsic subtypes of TNBC [47] for in vitro cytotoxicity experiments.

Fig. 1.

Fig. 1

EGFR is highly enriched in patient TNBC brain metastases and in vitro TNBC cell lines. a Tissue microarray of 13 patients with TNBC Brain Metastases, with scores prepared in duplicate. Pathologist score of membrane and cytoplasmic EGFR staining. Patient 13 (top inset) has highest H score of 300. Patient 9 (bottom inset) has membranous H-score of 240. b TNBC cell lines were surface stained by EGFR and underwent flow cytometry presented as mean fluorescence intensity (MFI). c Immunoblot of EGFR and actin of TNBC cell lines and EGFR KO MDA-MB-231

EGFR806 CAR T cells demonstrate effective in vitro tumor killing

We first designed a second-generation CART cell using the EGFR806 scFv to target the oncogenic EGFR expression on TNBC-BM patient tissue samples (Fig. 2). Mab806 is restricted to oncogenic amplification of EGFR [3638] and has been utilized for treating glioblastoma patients in clinical trials [46]. The Vh and Vl domain is followed by double mutated IgG spacer (IgG4EQ) to reduce Fc receptor recognition [45]. The endodomain consist of CD4 transmembrane domain, 4–1BB costimulatory domain and CD3ζ cytolytic domain (supplemental table S2). CAR expression was identified by flow cytometry using anti-human IgG, Fcγ fragment specific (109–546–008 Jackson Immuno Research, West Grove, Pennsylvania) (Fig. 2B,C). We transduced human CD4 cells with the EGFR806 lentivirus. We chose CD4 cells given their superior long-term persistence and recursive killing potential compared to CD8 T cells alone or combination CD4-CD8 T cells [48, 49].

We examined the in vitro killing of the EGFR806 CAR T cells against MDA-MB-231 over time course 24, 48, and 96 h at Effector to Tumor cell ratio of 1:1 (Fig. 3A). Most effective killing was found at 96 h at this E:T. Further, CAR T cells demonstrated statistically significant increase in T cell activation markers (Perforin and Granzyme B) after incubation with tumor cells compared with mock T cells(Fig. 3B).

Fig. 3.

Fig. 3

EGFR806 CAR T effectively targets TNBC cells in vitro and results in CAR activation. a Time course of killing 1:1 CAR T vs MDA-MB-231 at 24, 48, and 96 h. b CAR T activation markers Perforin and Granzyme B co-expression demonstrate increased activation after incubation with MD-MDA-231 cells. Data are represented as mean ± SD and were analyzed by student’s t-test. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001

Next, we evaluated specificity of EGFR806 CAR T cells cytolysis of against MDA-MB-231 scarmble control vs EGFR CRISPR-Cas9 KO MDA-MB-231cells (Fig. 4A, B). Cytolytic ability of EGFR806 CAR T cells was lost upon EGFR KO, confirming the specificity of the EGFR806 CAR. Next, we evaluated EGFR806 CAR killing using other cell lines in addition to MDA-MB-231 (MDA-MB-468 and HCC70 TNBC) cells by co-culturing tumor cells and CAR T cells (Fig. 4C). DAPI staining was our gating strategy to determine the percentage of dead cells by flow cytometry. Statistically significantly higher frequency of live cells was observed with EGFR806 CAR T cells with respect to Mock CD4 T cells as control for all the lines.

Fig. 4.

Fig. 4

EGFR806 CAR T is specific and kills multiple TNBC lines. a Second generation EGFR806 CAR does not kill EGFR806 KO Tumor cells. b Western Blot confirming KO of EGFR806 from MB 231 cells. c CAR T killing across 3 cell lines, incubated 1:1, at 96 h. Data are represented as mean ± SD and were analyzed by student’s t-test. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001

TNBC-BM tumor regression after ICT delivery of EGFR CAR T cells in vivo

To evaluate the tumor targeted killing of EGFR806 CAR T cells, NSG mice were intracranially injected with human TNBC cell line MDA-MB-231 cells transduced with firefly luciferase. Mice underwent bioluminescent imaging (BLI) one day prior to CAR T injection (Day 5 post tumor implantation) to quantify tumor burden, as seen in schema (Fig. 5A). Human CD4 CAR T cells were thawed from a lentivirus production that was frozen D7 after transduction, and flow cytometry was done to confirm viability and CAR expression. EGFR806 CAR T cells treated mice were eradicated of tumor without any BLI evidence of tumor recurrence Mice with BLI negative tumors underwent repeat BLI imaging to confirm absence of BLI signal. (Fig. 5B, C), statistically significant survival (Fig. 5D). All mice were euthanized at designated times. By H&E staining, there was no evidence of tumor in CAR T treated mice, with heavy disease burden and hemorrhage in control mice (Fig. 5E and supplementary Fig. s2).

Fig. 5.

Fig. 5

EGFR806 CAR T increases survival of TNBC-BM mice. a Schema: Lentivirus transduced MDA-MB-231 cells were implanted intracranially at Day 0. Tumor engraftment was confirmed by BLI followed by ICT delivered EGFR806 CAR T cells on Day 6. b Tumor burden monitored by BLI imaging. c Bioluminescent flux for each group and all mice is graphed. Differences in BLI are calculated by one-way ANOVA. d Mouse survival calculated by log-rank (Mantle-Cox). e Representative IHC of mouse treated in mock group and CAR T group

Discussion

TNBC brain metastases is incurable with standard therapy. Autologous T cell therapy holds promise for a potential curative treatment. However, tumor associated antigens (TAAs) are often heterogenous in solid tumors, limiting single TAA targeted CAR T cells due to antigen loss [50, 51]. Identifying highly enriched TAAs is critical for targeting the maximal tumor population. Previous studies have demonstrated EGFR expression in breast cancer brain metastases in approximately 40% of patients, although the TNBC population was not stated [29, 52]. Our data further supports EGFR as a viable target in TNBC brain metastases, as our TMA is specific for TNBC-BM and demonstrates high membrane and cytoplasmic EGFR expression in six of 13 patients. Because cytoplasmic TAA expression is generally hidden from CAR T cells, future strategies may include tyrosine kinase inhibitors to enhance TAA surface presentation and CAR killing [53].

To our knowledge, our study is the first to demonstrate EGFR mAb806 CAR T cell mediated killing of TNBC cells in vitro and in vivo. This is a clinically meaningful observation, as EGFR mAb806 targets oncogenic over-expressed EGFR with limited toxicity to endogenous EGFR, including brain astrocytes [46, 54]. Indeed, EGFR806 CAR T cells are currently undergoing ICT delivery in pediatric brain tumor patients (Brainchild-02 NCT03638167) with no obvious toxicity in early reports [41]. Thus, there is significant potential to rapidly translate this finding into early-phase clinical evaluation for TNBC-BM patients.

Our EGFR806 CAR had maximum killing at 96 h compared to earlier time point at 1:1 ratio. This is consistent with prior study evaluating mAb806 based EGFR CAR killing of glioblastoma cells at earlier time points at higher E:T ratio, and similar anti-tumor activity in vivo murine brain tumor models [46]. Future studies will be required to further optimize our CAR T construct, such as optimization of hinge region (IgG4EQ, CD8a, CD28, IgG hinge only), or additional co-stimulatory domain.

Further, our data support local–regional intracranial delivery of CAR T cells. There are two major routes of intracranial delivery: intracranial ventricular (ICV) and intracranial tumor (ICT). In the clinical setting, an ommaya reservoir is placed subcutaneously and attached with cathether placement in the ventricle (ICV) or tumor resection cavity (ICT). This way, CAR T cells can be injected into ommaya subcutaneously, and allow for repeated access of CSF for molecular correlative studies. Other benefits include CAR T bypass of blood–brain-barrier as well as mitigating risk of extra-cranial CAR T toxicity. Priceman et al. are using ICV delivery of HER2 CAR T cells for treatment of breast-to-brain metastasis [28] in an ongoing clinical trial (NCT03696030). Similarly, with respect to patients with TNBC brain metastases, the major delivery route will likely include ICV, as CAR T cells injected within the CSF will be able to circulate through CSF and targets multiple foci of disease.

Interestingly, there may be synergy in co-targeting EGFR and HER2 re-directed CAR T cells in breast to brain metastases based upon co-expression from patient brain metastases [55]. An “OR” gated CAR, such as a dual or single chain bispecific CAR T construct design may mitigate tumor associated antigen escape. However, this does increase risk of on-target off-tumor toxicity. Advanced logic gated “AND” gated CAR T design, such as Syn-Notch may help mitigate toxicity [56]. Further, other potential tumor associated antigen targets for TNBC CAR T cells have been reviewed elsewhere [57].

Our EGFR806 CAR T cells were produced from CD4 human T cells and demonstrate effective long term tumor eradication and mouse survival. It is believed that efficacy of adoptive cell therapy can be often attributed to CD8 T cells [58] and infusion of CD8 derived CD19 CAR T cells alone is sufficient for long-term B-cell eradication [58, 59]. However, recent clinical data demonstrate long-term (10 year+) persistence of CD19 CAR T cells is mostly CD4 based [60]. In support of this observation, CD4 (vs CD8) based IL13R∝2 redirected IgG4(EQ)-41BBζ CAR T cells in preclinical GBM models demonstrate improved long term persistence and recursive killing [49]. Further, in other in vitro and in vivo studies of CD4 CAR T cells demonstrate similar effectiveness in directly killing tumor cells [48, 61, 62], with less activation induced cell death compared to CD8 T cells [48, 49, 63].

Given the ongoing clinical safety reports in these patients, our data further supports clinical evaluation of intracranial delivered CD4 T derived EGFR806 CAR T to TNBC-BM patients. Future studies will help further interrogate TNBC brain tissue for other TAAs that may complement heterogenous EGFR expression, as well as TNBC-BM specific tumor micro-environment related immune-suppressive pathways that may be targeted in conjunction with EGFR.

Our study has limitations. Our TNBC cell lines tested demonstrate 100% surface EGFR expression, however the patient TNBC-BM tissue demonstrates more heterogenous membrane EGFR expression. Future studies of combination with EGFR tyrosine kinase inhibitors (TKI) may enhance surface EGFR expression and sensitize to EGFR806 CAR T cells. This is a promising strategy, as brain-penetrant EGFR TKIs are undergoing clinical study [64]. Future studies utilizing heterogenous tumor EGFR expression will help identify mechanisms of tumor resistance to EGFR806 CAR T. Furthermore, our TMA did not assess the EGFR expression in the primary tumor. It is not yet clear the association between primary breast tumor and TNBC-BM for EGFR expression with regards to EGFR enrichment once metastatic to brain.

Supplementary Material

Supplementary_Subham et al

Funding

This work was funded by the University of Kansas Cancer Center.

Footnotes

Supplementary Information The online version contains supplementary material available at https://doi.org/10.1007/s10549-022-06783-1.

Competing interests The authors have no relevant financial or non-financial interests to disclose.

Ethical approval This study was performed in line with the principles of the Declaration of Helsinki and its later amendments or comparable ethical standards. Ethical approval of this study was granted by the University of Kansas Medical Center Institutional Review Board (IRB) and IACUC on September 21, 2021.

Informed consent Informed consent was obtained from all University of Kansas Medical Center Biorepository Core Facility for all patient tissue presented in this study.

Consent to participate Informed consent was obtained from all individual participants included in the study.

Consent to publish The authors affirm that human research participants provided informed consent for de-identified immunohistochemistry in Fig. 1A.

Data availability

The original contributions presented in the study are included in the article/supplementary material. Further inquiries can be directed to the corresponding author. All data will be made available to interested parties upon reasonable request.

References

  • 1.Siegel RL, Miller KD, Fuchs HE, Jemal A (2022) Cancer statistics, 2022. CA 72:7–33. 10.3322/caac.21708 [DOI] [PubMed] [Google Scholar]
  • 2.Brosnan EM, Anders CK (2018) Understanding patterns of brain metastasis in breast cancer and designing rational therapeutic strategies. Ann Transl Med 6:163. 10.21037/atm.2018.04.35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, Diéras V, Hegg R, Im S-A, Shaw Wright G, Henschel V, Molinero L, Chui SY, Funke R, Husain A, Winer EP, Loi S, Emens LA (2018) Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med 379:2108–2121. 10.1056/NEJMoa1809615 [DOI] [PubMed] [Google Scholar]
  • 4.Garrido-Castro A, Lin N, Polyak K (2019) Insights into molecular classifications of triple-negative breast cancer: improving patient selection for treatment. Cancer Discov 9:176–198. 10.1158/2159-8290.Cd-18-1177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Badve SS, Penault-Llorca F, Reis-Filho JS, Deurloo R, Siziopikou KP, D’Arrigo C, Viale G (2021) Determining PD-L1 status in patients with triple-negative breast cancer: lessons learned from IMpassion130. JNCI. 10.1093/jnci/djab121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mittendorf EA, Philips AV, Meric-Bernstam F, Qiao N, Wu Y, Harrington S, Su X, Wang Y, Gonzalez-Angulo AM, Akcakanat A, Chawla A, Curran M, Hwu P, Sharma P, Litton JK, Molldrem JJ, Alatrash G (2014) PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res 2:361–370. 10.1158/2326-6066.Cir-13-0127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lin NU, Claus E, Sohl J, Razzak AR, Arnaout A, Winer EP (2008) Sites of distant recurrence and clinical outcomes in patients with metastatic triple-negative breast cancer: high incidence of central nervous system metastases. Cancer 113:2638–2645. 10.1002/cncr.23930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Niikura N, Hayashi N, Masuda N, Takashima S, Nakamura R, Watanabe K, Kanbayashi C, Ishida M, Hozumi Y, Tsuneizumi M, Kondo N, Naito Y, Honda Y, Matsui A, Fujisawa T, Oshitanai R, Yasojima H, Tokuda Y, Saji S, Iwata H (2014) Treatment outcomes and prognostic factors for patients with brain metastases from breast cancer of each subtype: a multicenter retrospective analysis. Breast Cancer Res Treat 147:103–112. 10.1007/s10549-014-3090-8 [DOI] [PubMed] [Google Scholar]
  • 9.Kim YJ, Kim JS, Kim IA (2018) Molecular subtype predicts incidence and prognosis of brain metastasis from breast cancer in SEER database. J Cancer Res Clin Oncol 144:1803–1816. 10.1007/s00432-018-2697-2 [DOI] [PubMed] [Google Scholar]
  • 10.Martin AM, Cagney DN, Catalano PJ, Warren LE, Bellon JR, Punglia RS, Claus EB, Lee EQ, Wen PY, Haas-Kogan DA, Alexander BM, Lin NU, Aizer AA (2017) Brain metastases in newly diagnosed breast cancer: a population-based study. JAMA Oncol 3:1069–1077. 10.1001/jamaoncol.2017.0001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Jaklevic MC (2021) CAR-T therapy is approved for non-hodgkin lymphoma. JAMA 325:1032–1032. 10.1001/jama.2021.3004 [DOI] [PubMed] [Google Scholar]
  • 12.Maus MV (2021) CD19 CAR T cells for adults with relapsed or refractory acute lymphoblastic leukaemia. Lancet 398:466–467. 10.1016/s0140-6736(21)01289-7 [DOI] [PubMed] [Google Scholar]
  • 13.Akhavan D, Alizadeh D, Wang D, Weist MR, Shepphird JK, Brown CE (2019) CAR T cells for brain tumors: lessons learned and road ahead. Immunol Rev 290:60–84. 10.1111/imr.12773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Choi J, Jung WH, Koo JS (2012) Clinicopathologic features of molecular subtypes of triple negative breast cancer based on immunohistochemical markers. Histol Histopathol 27:1481–1493. 10.14670/hh-27.1481 [DOI] [PubMed] [Google Scholar]
  • 15.Liu D, He J, Yuan Z, Wang S, Peng R, Shi Y, Teng X, Qin T (2012) EGFR expression correlates with decreased disease-free survival in triple-negative breast cancer: a retrospective analysis based on a tissue microarray. Med Oncol 29:401–405. 10.1007/s12032-011-9827-x [DOI] [PubMed] [Google Scholar]
  • 16.Martin V, Botta F, Zanellato E, Molinari F, Crippa S, Mazzucchelli L, Frattini M (2012) Molecular characterization of EGFR and EGFR-downstream pathways in triple negative breast carcinomas with basal like features. Histol Histopathol 27:785–792. 10.14670/hh-27.785 [DOI] [PubMed] [Google Scholar]
  • 17.Meseure D, Vacher S, Drak Alsibai K, Trassard M, Susini A, Le Ray C, Lerebours F, Le Scodan R, Spyratos F, Marc Guinebretiere J, Lidereau R, Bieche I (2012) Profiling of EGFR mRNA and protein expression in 471 breast cancers compared with 10 normal tissues: a candidate biomarker to predict EGFR inhibitor effectiveness. Int J Cancer 131:1009–1010. 10.1002/ijc.26434 [DOI] [PubMed] [Google Scholar]
  • 18.Park HS, Jang MH, Kim EJ, Kim HJ, Lee HJ, Kim YJ, Kim JH, Kang E, Kim SW, Kim IA, Park SY (2014) High EGFR gene copy number predicts poor outcome in triple-negative breast cancer. Mod Pathol 27:1212–1222. 10.1038/modpathol.2013.251 [DOI] [PubMed] [Google Scholar]
  • 19.Viale G, Rotmensz N, Maisonneuve P, Bottiglieri L, Montagna E, Luini A, Veronesi P, Intra M, Torrisi R, Cardillo A, Campagnoli E, Goldhirsch A, Colleoni M (2009) Invasive ductal carcinoma of the breast with the “triple-negative” phenotype: prognostic implications of EGFR immunoreactivity. Breast Cancer Res Treat 116:317–328. 10.1007/s10549-008-0206-z [DOI] [PubMed] [Google Scholar]
  • 20.Baselga J, Gómez P, Greil R, Braga S, Climent MA, Wardley AM, Kaufman B, Stemmer SM, Pêgo A, Chan A, Goeminne JC, Graas MP, Kennedy MJ, Ciruelos Gil EM, Schneeweiss A, Zubel A, Groos J, Melezínková H, Awada A (2013) Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J Clin Oncol 31:2586–2592. 10.1200/jco.2012.46.2408 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Carey LA, Rugo HS, Marcom PK, Mayer EL, Esteva FJ, Ma CX, Liu MC, Storniolo AM, Rimawi MF, Forero-Torres A, Wolff AC, Hobday TJ, Ivanova A, Chiu WK, Ferraro M, Burrows E, Bernard PS, Hoadley KA, Perou CM, Winer EP (2012) TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol 30:2615–2623. 10.1200/jco.2010.34.5579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Trédan O, Campone M, Jassem J, Vyzula R, Coudert B, Pacilio C, Prausova J, Hardy-Bessard AC, Arance A, Mukhopadhyay P, Aloe A, Roché H (2015) Ixabepilone alone or with cetuximab as first-line treatment for advanced/metastatic triple-negative breast cancer. Clin Breast Cancer 15:8–15. 10.1016/j.clbc.2014.07.007 [DOI] [PubMed] [Google Scholar]
  • 23.Fenn K, Maurer M, Lee SM, Crew KD, Trivedi MS, Accordino MK, Hershman DL, Kalinsky K (2020) Phase 1 study of erlotinib and metformin in metastatic triple-negative breast cancer. Clin Breast Cancer 20:80–86. 10.1016/j.clbc.2019.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Symonds L, Linden H, Gadi V, Korde L, Rodler E, Gralow J, Redman M, Baker K, Wu QV, Jenkins I, Kurland B, Garrison M, Smith J, Anderson J, Van Haelst C, Specht J (2019) Combined targeted therapies for first-line treatment of metastatic triple negative breast cancer-A Phase II trial of weekly nab-paclitaxel and bevacizumab followed by maintenance targeted therapy with bevacizumab and erlotinib. Clin Breast Cancer 19:e283–e296. 10.1016/j.clbc.2018.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA, Chae YK, Cristofanilli M, Gradishar WJ, Giles FJ (2017) Targeting Epidermal Growth Factor Receptor in triple negative breast cancer: new discoveries and practical insights for drug development. Cancer Treat Rev 53:111–119. 10.1016/j.ctrv.2016.12.010 [DOI] [PubMed] [Google Scholar]
  • 26.Nakai K, Hung MC, Yamaguchi H (2016) A perspective on anti-EGFR therapies targeting triple-negative breast cancer. Am J Cancer Res 6:1609–1623 [PMC free article] [PubMed] [Google Scholar]
  • 27.Song W, Hwang Y, Youngblood VM, Cook RS, Balko JM, Chen J, Brantley-Sieders DM (2017) Targeting EphA2 impairs cell cycle progression and growth of basal-like/triple-negative breast cancers. Oncogene 36:5620–5630. 10.1038/onc.2017.170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Priceman SJ, Tilakawardane D, Jeang B, Aguilar B, Murad JP, Park AK, Chang W-C, Ostberg JR, Neman J, Jandial R, Portnow J, Forman SJ, Brown CE (2018) Regional delivery of chimeric antigen receptor-engineered T cells effectively targets HER2(+) breast cancer metastasis to the brain. Clin Cancer Res 24:95–105. 10.1158/1078-0432.CCR-17-2041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bryan S, Witzel I, Borgmann K, Oliveira-Ferrer L (2021) Molecular mechanisms associated with brain metastases in HER2-positive and triple negative breast cancers. Cancers (Basel) 13:4137. 10.3390/cancers13164137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Grupka NL, Lear-Kaul KC, Kleinschmidt-DeMasters BK, Singh M (2004) Epidermal growth factor receptor status in breast cancer metastases to the central nervous system. Comparison with HER-2/neu status. Arch Pathol Lab Med 128:974–979. 10.5858/2004-128-974-egfrsi [DOI] [PubMed] [Google Scholar]
  • 31.Xia L, Zheng Z, Liu J-y, Chen Y-j, Ding J, Hu G-s, Hu Y-h, Liu S, Luo W-x, Xia N-s, Liu W (2021) Targeting triple-negative breast cancer with combination therapy of EGFR CAR T cells and CDK7 inhibition. Cancer Immunol Res 9:707–722. 10.1158/2326-6066.Cir-20-0405 [DOI] [PubMed] [Google Scholar]
  • 32.Xia L, Zheng ZZ, Liu JY, Chen YJ, Ding JC, Xia NS, Luo WX, Liu W (2020) EGFR-targeted CAR-T cells are potent and specific in suppressing triple-negative breast cancer both in vitro and in vivo. Clin Transl Immunol 9:e01135. 10.1002/cti2.1135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hübner J, Raschke M, Rütschle I, Gräßle S, Hasenberg T, Schirrmann K, Lorenz A, Schnurre S, Lauster R, Maschmeyer I, Steger-Hartmann T, Marx U (2018) Simultaneous evaluation of anti-EGFR-induced tumour and adverse skin effects in a microfluidic human 3D co-culture model. Sci Rep 8:15010. 10.1038/s41598-018-33462-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Laux I, Jain A, Singh S, Agus DB (2006) Epidermal growth factor receptor dimerization status determines skin toxicity to HER-kinase targeted therapies. Br J Cancer 94:85–92. 10.1038/sj.bjc.6602875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Choi BD, Yu X, Castano AP, Bouffard AA, Schmidts A, Larson RC, Bailey SR, Boroughs AC, Frigault MJ, Leick MB, Scarfò I, Cetrulo CL, Demehri S, Nahed BV, Cahill DP, Wakimoto H, Curry WT, Carter BS, Maus MV (2019) CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat Biotechnol 37:1049–1058. 10.1038/s41587-019-0192-1 [DOI] [PubMed] [Google Scholar]
  • 36.Jungbluth AA, Stockert E, Huang HJ, Collins VP, Coplan K, Iversen K, Kolb D, Johns TJ, Scott AM, Gullick WJ, Ritter G, Cohen L, Scanlan MJ, Cavenee WK, Old LJ (2003) A monoclonal antibody recognizing human cancers with amplification/overexpression of the human epidermal growth factor receptor. Proc Natl Acad Sci USA 100:639–644. 10.1073/pnas.232686499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Luwor RB, Johns TG, Murone C, Huang HJ, Cavenee WK, Ritter G, Old LJ, Burgess AW, Scott AM (2001) Monoclonal antibody 806 inhibits the growth of tumor xenografts expressing either the de2–7 or amplified epidermal growth factor receptor (EGFR) but not wild-type EGFR. Cancer Res 61:5355–5361 [PubMed] [Google Scholar]
  • 38.Panousis C, Rayzman VM, Johns TG, Renner C, Liu Z, Cart-wright G, Lee FT, Wang D, Gan H, Cao D, Kypridis A, Smyth FE, Brechbiel MW, Burgess AW, Old LJ, Scott AM (2005) Engineering and characterisation of chimeric monoclonal antibody 806 (ch806) for targeted immunotherapy of tumours expressing de2–7 EGFR or amplified EGFR. Br J Cancer 92:1069–1077. 10.1038/sj.bjc.6602470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Simon N, Antignani A, Sarnovsky R, Hewitt SM, FitzGerald D (2016) Targeting a cancer-specific epitope of the epidermal growth factor receptor in triple-negative breast cancer. J Natl Cancer Inst. 10.1093/jnci/djw028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Scott AM, Lee FT, Tebbutt N, Herbertson R, Gill SS, Liu Z, Skrinos E, Murone C, Saunder TH, Chappell B, Papenfuss AT, Poon AM, Hopkins W, Smyth FE, MacGregor D, Cher LM, Jungbluth AA, Ritter G, Brechbiel MW, Murphy R, Burgess AW, Hoffman EW, Johns TG, Old LJ (2007) A phase I clinical trial with monoclonal antibody ch806 targeting transitional state and mutant epidermal growth factor receptors. Proc Natl Acad Sci USA 104:4071–4076. 10.1073/pnas.0611693104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Vitanza N, Gust J, Wilson A, Huang W, Perez F, Wright J, Leary S, Cole B, Albert C, Pinto N, Orentas R, Jensen M, Park J (2020) IMMU-03. UPDATES ON BRAINCHILD-01, −02, AND −03: PHASE 1 LOCOREGIONAL CAR T CELL TRIALS TARGETING HER2, EGFR, AND B7–H3 FOR CHILDREN WITH RECURRENT CNS TUMORS AND DIPG. Neuro-Oncology 22:iii360–iii360. 10.1093/neuonc/noaa222.360 [DOI] [Google Scholar]
  • 42.Atlas P (2022) In:
  • 43.Huerta JJ, Diaz-Trelles R, Naves FJ, Llamosas MM, Del Valle ME, Vega JA (1996) Epidermal growth factor receptor in adult human dorsal root ganglia. Anat Embryol (Berl) 194:253–257. 10.1007/bf00187136 [DOI] [PubMed] [Google Scholar]
  • 44.Kornblum HI, Gall CM, Seroogy KB, Lauterborn JC (1995) A subpopulation of striatal gabaergic neurons expresses the epidermal growth factor receptor. Neuroscience 69:1025–1029. 10.1016/0306-4522(95)00392-v [DOI] [PubMed] [Google Scholar]
  • 45.Jonnalagadda M, Mardiros A, Urak R, Wang X, Hoffman LJ, Bernanke A, Chang WC, Bretzlaff W, Starr R, Priceman S, Ostberg JR, Forman SJ, Brown CE (2015) Chimeric antigen receptors with mutated IgG4 Fc spacer avoid fc receptor binding and improve T cell persistence and antitumor efficacy. Mol Ther 23:757–768. 10.1038/mt.2014.208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ravanpay AC, Gust J, Johnson AJ, Rolczynski LS, Cecchini M, Chang CA, Hoglund VJ, Mukherjee R, Vitanza NA, Orentas RJ, Jensen MC (2019) EGFR806-CAR T cells selectively target a tumor-restricted EGFR epitope in glioblastoma. Oncotarget 10:7080–7095. 10.18632/oncotarget.27389 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA (2011) Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 121:2750–2767. 10.1172/JCI45014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Agarwal S, Hanauer JDS, Frank AM, Riechert V, Thalheimer FB, Buchholz CJ (2020) In vivo generation of CAR T cells selectively in human CD4(+) lymphocytes. Mol Ther 28:1783–1794. 10.1016/j.ymthe.2020.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wang D, Aguilar B, Starr R, Alizadeh D, Brito A, Sarkissian A, Ostberg JR, Forman SJ, Brown CE (2018) Glioblastoma-targeted CD4+ CAR T cells mediate superior antitumor activity. JCI Insight. 10.1172/jci.insight.99048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, Ostberg JR, Blanchard MS, Kilpatrick J, Simpson J, Kurien A, Priceman SJ, Wang X, Harshbarger TL, D’Apuzzo M, Ressler JA, Jensen MC, Barish ME, Chen M, Portnow J, Forman SJ, Badie B (2016) Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med 375:2561–2569. 10.1056/NEJMoa1610497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.O’Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, Martinez-Lage M, Brem S, Maloney E, Shen A, Isaacs R, Mohan S, Plesa G, Lacey SF, Navenot JM, Zheng Z, Levine BL, Okada H, June CH, Brogdon JL, Maus MV (2017) A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 10.1126/scitranslmed.aaa0984 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sirkisoon SR, Carpenter RL, Rimkus T, Miller L, Metheny-Barlow L, Lo H-W (2016) EGFR and HER2 signaling in breast cancer brain metastasis. Front Biosci (Elite Ed) 8:245–263. 10.2741/E765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Heczey A (2019) Alliance of the titans: an effective combination of a TKI with CAR T cells. Mol Ther 27:1348–1349. 10.1016/j.ymthe.2019.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Simon N, Antignani A, Sarnovsky R, Hewitt SM, FitzGerald D (2016) Targeting a cancer-specific epitope of the epidermal growth factor receptor in triple-negative breast cancer. JNCI. 10.1093/jnci/djw028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Guo P, Pu T, Chen S, Qiu Y, Zhong X, Zheng H, Chen L, Bu H, Ye F (2017) Breast cancers with EGFR and HER2 co-amplification favor distant metastasis and poor clinical outcome. Oncol Lett 14:6562–6570. 10.3892/ol.2017.7051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Choe JH, Watchmaker PB, Simic MS, Gilbert RD, Li AW, Krasnow NA, Downey KM, Yu W, Carrera DA, Celli A, Cho J, Bri-ones JD, Duecker JM, Goretsky YE, Dannenfelser R, Cardarelli L, Troyanskaya O, Sidhu SS, Roybal KT, Okada H, Lim WA (2021) SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med. 10.1126/scitranslmed.abe7378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Dees S, Ganesan R, Singh S, Grewal IS (2020) Emerging CAR-T cell therapy for the treatment of triple-negative breast cancer. Mol Cancer Ther 19:2409–2421. 10.1158/1535-7163.Mct-20-0385 [DOI] [PubMed] [Google Scholar]
  • 58.Terakura S, Yamamoto TN, Gardner RA, Turtle CJ, Jensen MC, Riddell SR (2012) Generation of CD19-chimeric antigen receptor modified CD8+ T cells derived from virus-specific central memory T cells. Blood 119:72–82. 10.1182/blood-2011-07-366419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang X, Naranjo A, Brown CE, Bautista C, Wong CW, Chang WC, Aguilar B, Ostberg JR, Riddell SR, Forman SJ, Jensen MC (2012) Phenotypic and functional attributes of lentivirus-modified CD19-specific human CD8+ central memory T cells manufactured at clinical scale. J Immunother 35:689–701. 10.1097/CJI.0b013e318270dec7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, Gao P, Bandyopadhyay S, Sun H, Zhao Z, Lundh S, Pruteanu-Malinici I, Nobles CL, Maji S, Frey NV, Gill SI, Tian L, Kulikovskaya I, Gupta M, Ambrose DE, Davis MM, Fraietta JA, Brogdon JL, Young RM, Chew A, Levine BL, Siegel DL, Alanio C, Wherry EJ, Bushman FD, Lacey SF, Tan K, June CH (2022) Decade-long leukaemia remissions with persistence of CD4(+) CAR T cells. Nature 602:503–509. 10.1038/s41586-021-04390-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Xhangolli I, Dura B, Lee G, Kim D, Xiao Y, Fan R (2019) Single-cell analysis of CAR-T cell activation reveals A mixed TH1/TH2 response independent of differentiation. Genom Proteom Bioin-form 17:129–139. 10.1016/j.gpb.2019.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yang Y, Kohler ME, Chien CD, Sauter CT, Jacoby E, Yan C, Hu Y, Wanhainen K, Qin H, Fry TJ (2017) TCR engagement negatively affects CD8 but not CD4 CAR T cell expansion and leukemic clearance. Sci Transl Med. 10.1126/scitranslmed.aag1209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Liadi I, Singh H, Romain G, Rey-Villamizar N, Merouane A, Adolacion JR, Kebriaei P, Huls H, Qiu P, Roysam B, Cooper LJ, Varadarajan N (2015) Individual motile CD4(+) T cells can participate in efficient multikilling through conjugation to multiple tumor cells. Cancer Immunol Res 3:473–482. 10.1158/2326-6066.Cir-14-0195 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tsang JE, Urner LM, Kim G, Chow K, Baufeld L, Faull K, Cloughesy TF, Clark PM, Jung ME, Nathanson DA (2020) Development of a potent brain-penetrant EGFR tyrosine kinase inhibitor against malignant brain tumors. ACS Med Chem Lett 11:1799–1809. 10.1021/acsmedchemlett.9b00599 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary_Subham et al

Data Availability Statement

The original contributions presented in the study are included in the article/supplementary material. Further inquiries can be directed to the corresponding author. All data will be made available to interested parties upon reasonable request.

RESOURCES