Quick viewing(Text Mode)

Antibody Drug Conjugates: Future Directions in Clinical and Translational Strategies to 2 Improve the Therapeutic Index

Antibody Drug Conjugates: Future Directions in Clinical and Translational Strategies to 2 Improve the Therapeutic Index

Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1

1 Antibody Drug Conjugates: Future Directions in Clinical and Translational Strategies to 2 Improve the Therapeutic Index

3

4 Authors: Steven Coats1, Marna Williams1, Benjamin Kebble1, Rakesh Dixit1, Leo Tseng1, Nai- 5 Shun Yao1, David A. Tice1, and Jean-Charles Soria1,2

6

7 Affiliations: 1AstraZeneca, Gaithersburg, MD, USA. 2University Paris-Sud, Orsay, France.

8

9 Running title: Advances in Antibody Drug Conjugate Clinical Development

10

11 Corresponding Author: Steven Coats

12 Research and Development Oncology

13 AstraZeneca

14 1 MedImmune Way

15 Gaithersburg, MD 20878

16 Email: [email protected]

17

18 DISCLOSURE OF POTENTIAL CONFLICT OF INTEREST

19 All authors are employees of AstraZeneca and hold stock/stock options in AstraZeneca. Dr. 20 Soria also holds stock/stock options in Gritstone. Over the last 5 years, Dr. Soria has received 21 consultancy fees from AstraZeneca, Astex, Clovis, GSK, GamaMabs, Lilly, MSD, Mission 22 Therapeutics, Merus, , PharmaMar, Pierre Fabre, Roche/Genentech, Sanofi, Servier, 23 Symphogen, and Takeda.

24

25 26

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

2

27 ABSTRACT

28 Since the first approval of (Mylotarg; CD33 targeted), 2 additional 29 antibody drug conjugates (ADCs)— (Adcetris; CD30 targeted) and 30 (Besponsa; CD22 targeted)—have been approved for hematologic 31 cancers and 1 ADC, emtansine (Kadcyla; HER2 targeted), has been approved to 32 treat . Despite a clear clinical benefit being demonstrated for all 4 approved ADCs, 33 the toxicity profiles are comparable to those of standard-of-care chemotherapeutics, with dose- 34 limiting toxicities associated with the mechanism of activity of the cytotoxic warhead. However, 35 the enthusiasm to develop ADCs has not been dampened; approximately 80 ADCs are in 36 clinical development in nearly 600 clinical trials, and 2 to 3 novel ADCs are likely to be approved 37 within the next few years. While the promise of a more targeted with less toxicity 38 has not yet been realized with ADCs, improvements in technology combined with a wealth of 39 clinical data are helping to shape the future development of ADCs. In this review we discuss the 40 clinical and translational strategies associated with improving the therapeutic index for ADCs.

41

42

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

3

43 Introduction

44 Antibody drug conjugates (ADCs) were initially designed to leverage the exquisite specificity of 45 antibodies to deliver targeted potent chemotherapeutic agents with the intention of improving 46 the therapeutic index (the ratio between the toxic dose and the dose at which the drug becomes 47 effective; Figure 1) (1, 2). Unfortunately, the greatest challenge to date for developing ADCs is 48 a therapeutic index far narrower than expected (3-5). Of approximately 55 traditional ADCs for 49 which clinical development has been halted, we estimate that at least 23 have been 50 discontinued due to a poor therapeutic index; however, this is likely a conservative estimate 51 based on the availability of clinical data. A narrow therapeutic window limits the dose that can 52 be achieved, often resulting in toxic effects occurring before an ADC reaches its maximally 53 efficacious dose. Furthermore, these toxicities limit the number of dosing cycles that patients 54 can tolerate and often result in skipped doses, dose reductions, or study discontinuations (6, 7). 55 56 In this review we discuss clinical and translational strategies to improve the therapeutic index of 57 ADCs that are based on the latest clinical efficacy and safety data with next-generation 58 antibodies and warheads currently in development. While technology plays a crucial role in 59 expanding the therapeutic index of ADCs, we refer readers to several excellent reviews that 60 cover novel advancements in antibody, linker, and warhead technologies in significant depth (2, 61 3, 8, 9)

62 Overview of ADCs in Clinical Development

63 Four ADCs have been approved over the last 20 years (Figure 2A)(2). The first ADC approved 64 for clinical use was gemtuzumab ozogamicin (Mylotarg; CD33 targeted) for relapsed acute 65 myeloid in 2000 (10). In 2010, gemtuzumab ozogamicin was withdrawn from the US 66 market when a confirmatory trial showed that it was associated with a greater rate of fatal 67 toxicities vs standard-of-care chemotherapy (5.8% vs 0.8%) (10, 11). In 2017, gemtuzumab 68 ozogamicin was reapproved for relapsed/refractory after a phase 3 trial 69 with a fractionated dosing schedule lowered the peak serum concentration and improved the 70 safety profile, with a complete response rate of 26% (12). These clinical data demonstrate the 71 importance of understanding the relationship between the exposure, safety, and efficacy of 72 ADCs in clinical development.

73 Other ADCs that have been approved are brentuximab vedotin (Adcetris; CD30 targeted) (13) 74 and inotuzumab ozogamicin (Besponsa; CD22 targeted) (14), which were approved for

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

4

75 hematologic malignancies, and (Kadcyla; HER2 targeted), which was 76 approved for breast cancer (15). Across phase 2 and 3 studies, response rates were 77 significantly higher in patients treated with ADCs than in those treated with standard intensive 78 chemotherapy (14, 16-18).

79 Clear clinical benefits have been demonstrated with all 4 approved ADCs; however, each has 80 reported toxicity profiles that are specific to its cytotoxic warhead and, therefore, they cannot be 81 differentiated from standard-of-care (13-15) in terms of safety. Regardless of 82 the obstacles, there is intense interest in developing ADCs—approximately 80 ADC candidates 83 are reportedly in clinical development, with nearly 600 clinical trials ongoing—and it is likely that 84 several new ADCs will be approved over the next few years (Figure 2A) (19) led by the recent 85 Biologics License Application filing for (CD79b targeted) in 86 relapsed/refractory DLBCL. Although ADCs have not yet delivered on the promise of a more- 87 targeted chemotherapy with an improved toxicity profile, new strategies may prove crucial to 88 improving the therapeutic index of ADCs (4, 20, 21). These strategies include the use of 89 warheads with lower potencies and alternative mechanisms of activity as described below.

90 Two examples of ADCs in clinical development that use warheads that inhibit topoisomerase I 91 activity include targeting HER2 in breast and gastric cancers and 92 targeting Trop2 in breast and lung cancers (22, 23). A Biologics License 93 Application has been filed for sacituzumab govitecan for metastatic triple-negative breast 94 cancer, and trastuzumab deruxtecan is currently in multiple late-stage pivotal clinical trials. The 95 clinical data for trastuzumab deruxtecan from an ongoing phase 1 study in HER2-high 96 metastatic breast cancer (post trastuzumab emtansine) showed an ORR of 55% with median 97 progression-free survival not reached (Table 1). Updated recent data have shown a median 98 duration of response of 20.7 months, which compares favorably with trastuzumab emtansine, 99 which, in a pivotal study in HER2-high metastatic breast cancer, showed an ORR of 43.6%, a 100 median progression-free survival of 9.6 months, and a median duration of response of 12.6 101 months (22). In a phase 1 trial in third-line triple-negative breast cancer, sacituzumab govitecan 102 demonstrated an ORR of 31% and a median progression-free survival of 5.5 months (Table 1). 103 In this trial, sacituzumab govitecan was dosed at 10 mg/kg on days 1 and 8 every 21 days and 104 showed improved tolerability compared with other ADCs targeting Trop2 such as PF-06664178, 105 which had a maximum tolerated dose of 2.4 mg/kg, showed limited efficacy, and was terminated 106 due to high toxicity (23). 107

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

5

108 The results from this phase 1 trial with sacituzumab govitecan provide an example of the 109 importance of matching the right drug to the right target for the right patient. Even when 110 comparing ADCs that use the same antibody against the same target in a similar patient 111 population, trastuzumab emtansine and trastuzumab deruxtecan have demonstrated clinical 112 activity, whereas a trastuzumab tesirine conjugate (ADCT-502) was recently discontinued due 113 to a narrow therapeutic index (24). HER2 is known to be expressed in several normal tissues 114 such as in the lung and the gastrointestinal tract (25). This creates 2 potential problems for an 115 ADC. First, the normal expression of the creates a sink for the ADC that must be 116 overcome to maximize exposure to the tumor (26, 27). Given the high potency of the tesirine 117 payload, doses sufficient to overcome the HER2 normal tissue sink might not be achievable. 118 Second, the normal expression of the antigen can result in on-target toxicity. In the case of 119 trastuzumab tesirine, pulmonary edema, a known toxicity of pyrrolobenzodiazepines (28), may 120 have been exacerbated by the expression of HER2 in lung tissues. While general 121 characteristics of an ADC target, such as tumor-to-normal expression ratios and internalization 122 kinetics, may be considered, both the HER2 and Trop2 examples provide evidence that 123 achieving clinical success with an ADC may depend on matching the technology and the target. 124 125 The non–target-mediated uptake of the cytotoxic drug into normal tissues remains a challenge 126 with ADCs, thus limiting their therapeutic index. Although the immunoglobulin G (IgG) portion of 127 the ADC is important for maintaining a long half-life, binding to target, and internalizing drug into 128 tumor cells, its large size presents a physical barrier to efficient extravasation across blood 129 vessel walls and diffusion through tumors (29). This has prompted a significant effort to explore 130 alternative formats to traditional IgGs, including antibody fragments, alternative scaffolds, 131 natural ligands, and small molecules (30). Three drug conjugates using smaller targeting 132 domains have now entered the clinic. PEN-221 is a Pentarin (Tarveda Therapeutics) peptide 133 targeting the somatostatin receptor 2 conjugated to DM1 (clinicaltrials.gov identifier: 134 NCT02936323). PEN-866 is a small-molecule HSP90-binding ligand conjugated to SN38 (31) 135 (clinicaltrials.gov identifier: NCT03221400). BT-1718 is a bicyclic peptide targeting matrix 136 metalloprotease 14 and is conjugated to DM1 (32, 33) (clinicaltrials.gov identifier: 137 NCT03486730). Although small formats have been shown to extravasate and diffuse through 138 tissue faster than full-length IgG, the longer half-life of an IgG allows for greater absolute drug 139 accumulation into tumors over time (34, 35). However, the faster clearance may improve the 140 therapeutic index because the biodistribution is fundamentally changed, thereby altering normal

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

6

141 tissue exposure to both intact conjugate and released drug. It remains to be seen whether these 142 technologies will offer any improvement in the clinical therapeutic index. 143 144 145

146 Emerging Clinical and Translational Approaches

147 Maximizing the therapeutic index through clinical and translational strategies is central to the 148 future success of ADCs. There are several approaches that may be considered, including but 149 not limited to alteration of dosing regimen and use of biomarkers to optimize patient selection, 150 capture response signals early, and inform potential combination therapies. These approaches 151 are central to maximizing the therapeutic index and providing a personalized approach to ADC 152 therapeutic development.

153 Clinical dosing schedule 154 One approach to overcoming a narrow therapeutic index involves changing dosing schedules 155 through fractionated dosing. A fractionated dosing schedule may help maintain or increase dose 156 intensity—which is considered a major driver of anticancer activity—while reducing the peak 157 concentration. This approach has the potential to reduce the maximum serum concentration– 158 driven toxicities and prolong exposure, thereby ensuring that a greater number of cancer cells 159 enter the cell cycle and are exposed to drug. This has proven effective in traditional 160 chemotherapeutics, such as in adjuvant breast cancer (36, 37). Furthermore, the success of 161 fractionated dosing schedules with gemtuzumab ozogamicin or inotuzumab ozogamicin 162 suggests that the same approach can be used with other ADCs . Indeed, a preclinical study of 163 ADCs with pyrrolobenzodiazepine (PBD) warheads demonstrated that the in vivo efficacy and 164 area under the concentration curve were similar regardless of whether the ADC was delivered 165 as a single dose or as fractionated weekly doses, but that fractionated dosing reduced the 166 plasma concentration of the drug and therefore reduced maximum serum concentration–driven 167 toxicities (38). 168 169 Biodistribution studies 170 Biodistribution studies can help define target density beyond tumor cells and have the potential 171 to inform target-mediated and nontarget mediated toxicity. Biodistribution studies in humans 172 based on imaging analysis may be required, because target expression in animal models may

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

7

173 not reflect distribution in humans (39, 40). Indeed, a recent study of positron emission 174 tomography (PET) imaging with zirconium-89 labeled trastuzumab to assess HER2 status 175 demonstrated substantial heterogeneity in HER2 expression in metastatic lesions within the 176 same patients (41, 42). Combining the imaging analysis with fludeoxyglucose F 18–labeled 177 PET/computed tomography imaging enabled prediction of which patients would benefit from 178 treatment with the HER2 ADC (41). Unfortunately, preclinical models have not reflected the 179 heterogeneity of target expression that is seen across multiple metastatic lesions in the 180 populations of patients with relapsed and refractory disease who are frequently treated with 181 ADCs. Imaging analysis was also used to understand tumor distribution of ADCs in a study that 182 demonstrated significant differences in tumor uptake between an unconjugated Lewis Y 183 and the same Lewis Y monoclonal antibody conjugated to 184 (43, 44). However, different dose ranges were applied for naked antibody vs. antibody 185 conjugate, and nonlinear pharmacokinetics were observed, complicating data interpretation. 186 Nevertheless, the results suggest that the process of conjugating a warhead onto an antibody 187 may potentially alter the biophysical properties of the antibody, which could impact its 188 biodistribution profile.These imaging examples underscore an opportunity to more fully 189 understand the target expression profile in patients before they are treated with ADCs and to 190 determine the potential impact on the biodistribution properties of an antibody following 191 conjugation to a warhead. 192 193 Biomarkers to optimize patient selection 194 Patient-selection strategies with ADCs have previously focused primarily on target receptor 195 expression on tumor cells; however, a more comprehensive strategy that includes markers 196 linked to the mechanism of action of ADCs can be used to improve the likelihood of success 197 (Figure 3). One component of potential sensitivity to ADCs is patient response to warheads 198 linked to the monoclonal antibody. Biomarkers associated with warhead sensitivity could provide 199 an opportunity to improve the therapeutic index by observing responses at lower doses of 200 ADCs, which, in turn, may broaden the therapeutic index. Although these types of sensitivity 201 markers have been identified for some chemotherapies (45-48), they have not been used for 202 patient selection; however, the more targeted approaches of ADCs may enable patient selection 203 strategies based on warhead sensitivity profiles. 204 205 Biomarkers of DNA damage response have been used for patient selection for DNA damage 206 repair inhibitors such as poly ADP (adenosine diphosphate)-ribose polymerase (PARP)

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

8

207 inhibitors (49-56). Similarly, with warheads that induce DNA damage, such as topoisomerase 208 inhibitors (TOPi) and PBD dimers, patients with aberrations in DNA damage repair pathways 209 may have improved responses and, potentially, a broader therapeutic window. Selective 210 knockdown and knockout of genes involved in DNA damage response (BRCA1 and BRCA2) 211 have been shown to sensitize killing by PBD dimers (57). Furthermore, an ADC conjugated to 212 PBD demonstrated improved potency in xenografts with mutations in BRCA genes compared 213 with wild-type xenografts, providing proof of concept for candidate PBD-response markers for 214 clinical evaluation (57). Specific knockouts, knockdowns, and mutations in DNA damage 215 response (DDR) genes and/or genes potentially involved in the regulation of DDR have been 216 shown to confer sensitivity of tumor cells to TOPi (58-60) and PBDs (61). Interestingly, while 217 some of the sensitivity genes are shared (such as BRCA1, BRCA2, ATR, and FANCD2), others 218 differ which may reflect differences in the mechanism of each specific warhead that could 219 potentially contribute to differences in patient response.

220 While warhead sensitivity biomarkers have not been widely used for enrichment or pre-selection 221 of patients, aberrations in DDR pathway genes can be evaluated through analysis of tissue 222 biopsies as well as circulating tumor DNA (ctDNA) where DDR genes are included in several 223 genomics panels qualified for clinical studies (Clinical Laboratory Improvement Amendments 224 certified). Evaluation of ctDNA is less invasive for patients, and studies have shown 225 concordance of genomic profiles in ctDNA and tumor tissue (62); however, similar concordance 226 analyses will be needed in clinical studies to develop DDR genes as candidate predictive 227 biomarkers of response. In addition to sensitivity to DDR, other factors may impact warhead 228 sensitivity for DNA damaging agents; for example, for topoisomerase inhibitors, expression of 229 topoisomerases in target tumor cells may also impact clinical activity (63).

230 Compared to biomarkers for DNA-damaging agents, for microtubule inhibitors, tubulin isoforms 231 and a high proliferation index may sensitize patients to response. In preclinical studies, 232 decreases were preferentially observed in highly proliferating B cells (Ki-67+ CD20+ 233 lymphocytes) compared with nonproliferating B cells (Ki-67− CD20+ lymphocytes) after anti- 234 CD22-MMAE and anti-CD79b-MMAE treatment (64). 235

236 Biomarkers to capture response signals early and monitor the duration and depth of 237 response 238 Another factor central to the engineering of successful ADCs is the ability to capture response 239 signals early and to effectively monitor the depth and duration of response. This can be

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

9

240 especially important when attempting to optimize therapeutic index when testing new dosing 241 regimens. ctDNA can provide a noninvasive means of monitoring both longitudinal changes in 242 tumor burden and patients’ mutational profiles. While ctDNA levels have been shown to 243 associate with the response to cancer immunotherapies (65), the effects of ADCs on ctDNA and 244 associations with response have not been reported and could provide complementary 245 information to support and better understand clinical activity. ADCs have been shown to be 246 effective in hematology-oncology indications, including 3 of the 4 approved ADCs (gemtuzumab 247 ozogamicin, brentuximab vedotin, and inotuzumab ozogamicin) (10-14); establishing a means of 248 monitoring the changes in tumor burden in without invasive sampling could help 249 make development in these indications more efficient and less burdensome for patients. 250 251 Biomarkers to inform combination studies 252 Combining ADCs with immune checkpoint inhibitors, T-cell agonists, and other agents that 253 affect immunoresponse has the potential to reverse many of the evasive strategies that tumors 254 use to circumvent immunosurveillance. Currently, approximately 36 trials with 20 individual 255 ADCs in combination with immuno-oncology (IO) therapies are ongoing, most of which are 256 checkpoint inhibitors (Figure 2B). Early clinical data are available for 2 trials (66, 67). For 257 mirvetuximab in combination with , data indicate that responses are similar to 258 those with monotherapy; however, firm conclusions cannot be made at this time due to limited 259 data (66). The combination of ado-trastuzumab emtansine (T-DM1) and was 260 investigated in HER2+ metastatic breast cancer; although no clinically significant benefit was 261 observed with the combination in the intent-to-treat population, there was a trend towards 262 clinical benefit in biomarker-selected subsets of patients (67). 263 264 Preclinical evidence indicates that ADCs can induce immunogenic cell death (68, 69) and 265 provide synergistic antitumor activity when combined with IO agents (70-72). Treatment with 266 ADCs in syngeneic mouse models has been shown to lead to increased infiltration of actively 267 proliferating cytotoxic T lymphocytes and antigen-presenting cells in the tumor 268 microenvironment (TME) (71). Furthermore, infiltration of T cells has been observed in tumor 269 biopsy specimens from patients after treatment with T-DM1 (70). 270 271 The rationale that combinations of ADCs and IO agents will improve clinical activity centers on 272 the hypothesis that ADC treatment will alter the inflammatory milieu of tumor tissue, and 273 patients with antitumor immune responses will be more likely to benefit from combination

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

10

274 therapy. To assess the potential benefit of combining ADCs with IO agents, biomarkers can be 275 used to evaluate the TME before and after ADC monotherapy. Monitoring changes in the TME 276 after monotherapy can help determine whether markers predictive of response are upregulated 277 such as infiltration of T cells (73), elevated programmed death receptor 1 ligand (PD-L1) (74, 278 75), interferon-γ(IFN-γ), and IFN-γ–inducible factors (76) involved in T-cell regulation and 279 recruitment of immune cells into the TME. Tumor mutational burden and changes in T-cell 280 receptor diversity and clonal expansion can also be evaluated to determine if tumor-specific 281 neoantigens are being released by ADC treatment. These changes could help determine 282 whether ADCs can change “cold” TME to immunologically “warm/hot” TME. In parallel, 283 biomarkers associated with activation of immune responses could be evaluated in peripheral 284 blood, such as increases in proliferating (Ki-67+) T cells and markers of immunogenic cell death.

285 Evaluation of these changes in peripheral blood and tumor tissue may provide a better 286 understanding of the potential to improve ADC activity through combination treatment and help 287 prioritize disease indications with the highest likelihood of success. Furthermore, these 288 evaluations may be informative when considering dose adjustments to maximize the therapeutic 289 index. Patients not demonstrating changes in the TME indicating a response to checkpoint 290 inhibitors and/or markers of immunogenic cell death may be considered for dose adjustments 291 and/or other combination strategies (eg, T-cell agonists, oncolytic virus, or tumor vaccines).

292

293 Conclusions

294 With more than 80 compounds in various stages of clinical development, ADCs continue to be a 295 cancer treatment modality with significant investment and the ambition to selectively deliver 296 cytotoxic agents to cancer cells through specific binding of an antibody to cancer-selective 297 targets. Although clinical gaps remain regarding the optimal application of ADCs in oncology, 298 the study of these agents in a variety of settings is harnessing novel technologies and 299 leveraging translational medicine to maximize the therapeutic index of these agents. 300 301 Clinical development strategies will include alternative dosing schedules and cutting-edge 302 translational medicine to optimize patient selection, capture response signals early, match 303 biomarkers to warhead mechanisms of action, and evaluate potential combination therapies to 304 maximize the therapeutic index of ADCs. By incorporating these novel technologies and

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

11

305 biomarker selection strategies, ADCs will be well positioned to provide clinical benefit to a much 306 broader patient population. 307

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

12

308

309

310 SUMMARY OF AUTHOR CONTRIBUTIONS

311 All authors contributed to the concept, development, and review of all stages of this manuscript. 312

313 ACKNOWLEDGMENTS

314 Supported by AstraZeneca. Medical writing support was provided by Emily Weikum, PhD, of 315 SciMentum, Inc (Nucleus Global), funded by AstraZeneca, under the authors’ conceptual 316 direction and based on feedback from the authors.

317 318 319 320

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

13

321 References

322 1. Schrama D, Reisfeld RA, Becker JC. Antibody targeted drugs as cancer therapeutics. Nat

323 Rev Drug Discov 2006;5(2):147-59.

324 2. Dan N, Setua S, Kashyap VK, Khan S, Jaggi M, Yallapu MM, et al. Antibody-drug conjugates

325 for cancer therapy: Chemistry to clinical implications. Pharmaceuticals (Basel)

326 2018;11(2):10.3390/ph11020032.

327 3. Beck A, Goetsch L, Dumontet C, Corvaia N. Strategies and challenges for the next

328 generation of antibody-drug conjugates. Nat Rev Drug Discov 2017;16(5):315-37.

329 4. Vankemmelbeke M, Durrant L. Third-generation antibody drug conjugates for cancer therapy-

330 -a balancing act. Ther Deliv 2016;7(3):141-4.

331 5. Tolcher AW. Antibody drug conjugates: Lessons from 20 years of clinical experience. Ann

332 Oncol 2016;27(12):2168-72.

333 6. Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of

334 antibody-drug conjugates. MAbs 2016;8(4):659-71.

335 7. de Goeij BE, Lambert JM. New developments for antibody-drug conjugate-based therapeutic

336 approaches. Curr Opin Immunol 2016;40:14-23.

337 8. Jackson D, Stover D. Using the lessons learned from the clinic to improve the preclinical

338 development of antibody drug conjugates. Pharm Res 2015;32(11):3458-69.

339 9. Prabhu S, Boswell CA, Leipold D, Khawli LA, Li D, Lu D, et al. Antibody delivery of drugs and

340 radionuclides: Factors influencing clinical pharmacology. Ther Deliv 2011;2(6):769-91.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

14

341 10. Godwin CD, Gale RP, Walter RB. Gemtuzumab ozogamicin in acute myeloid leukemia.

342 Leukemia 2017;31(9):1855-68.

343 11. Petersdorf S, Kopecky K, Stuart RK, Larson RA, Nevill TJ, Stenke L, et al. Preliminary

344 results of southwest oncology group study S0106: An international intergroup phase 3

345 randomized trial comparing the addition of gemtuzumab ozogamicin to standard induction

346 therapy versus standard induction therapy followed by a second randomization to post-

347 consolidation gemtuzumab ozogamicin versus no additional therapy for previously untreated

348 acute myeloid leukemia. Blood 2009;114(22):790.

349 12. Jen EY, Ko CW, Lee JE, Del Valle PL, Aydanian A, Jewell C, et al. FDA approval:

350 Gemtuzumab ozogamicin for the treatment of adults with newly diagnosed CD33-positive acute

351 myeloid leukemia. Clin Cancer Res 2018;24(14):3242-6.

352 13. Yi JH, Kim SJ, Kim WS. Brentuximab vedotin: Clinical updates and practical guidance.

353 Blood Res 2017;52(4):243-53.

354 14. Lamb YN. Inotuzumab ozogamicin: First global approval. Drugs 2017;77(14):1603-10.

355 15. Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, et al. Trastuzumab emtansine

356 for HER2-positive advanced breast cancer. N Engl J Med 2012;367(19):1783-91.

357 16. Kantarjian HM, DeAngelo DJ, Stelljes M, Martinelli G, Liedtke M, Stock W, et al. Inotuzumab

358 ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med

359 2016;375(8):740-53.

360 17. Senter PD, Sievers EL. The discovery and development of brentuximab vedotin for use in

361 relapsed hodgkin and systemic anaplastic large cell lymphoma. Nat Biotechnol

362 2012;30(7):631-7.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

15

363 18. Younes A, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, et al. Results of a

364 pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory hodgkin's

365 lymphoma. J Clin Oncol 2012;30(18):2183-9.

366 19. Clinical trials database, website available at https://Www.clinicaltrials.gov/

367 [https://clinicaltrials.gov/ct2/home].

368 20. Chen H, Lin Z, Arnst KE, Miller DD, Li W. Tubulin inhibitor-based antibody-drug conjugates

369 for cancer therapy. Molecules 2017;22(8):10.3390/molecules22081281.

370 21. Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, et al. DS-8201a, A novel

371 HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising

372 antitumor efficacy with differentiation from T-DM1. Clin Cancer Res 2016;22(20):5097-108.

373 22. FY2018 R&D day

374 [https://www.daiichisankyo.com/media_investors/investor_relations/ir_calendar/files/005403/DS

375 _R&D%20Day%202018_E.pdf]. 2018.

376 23. King GT, Eaton KD, Beagle BR, Zopf CJ, Wong GY, Krupka HI, et al. A phase 1, dose-

377 escalation study of PF-06664178, an anti-trop-2/Aur0101 antibody-drug conjugate in patients

378 with advanced or metastatic solid tumors. Invest New Drugs 2018;36(5):836-47.

379 24. Study of ADCT-502 in patients with advanced solid tumors with HER2 expression -

380 clinicaltrials.gov [https://clinicaltrials.gov/ct2/show/NCT03125200]. 2018.

381 25. Press MF, Cordon-Cardo C, Slamon DJ. Expression of the HER-2/neu proto-oncogene in

382 normal human adult and fetal tissues. Oncogene 1990;5(7):953-62.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

16

383 26. Tabrizi MA, Tseng CM, Roskos LK. Elimination mechanisms of therapeutic monoclonal

384 antibodies. Drug Discov Today 2006;11(1-2):81-8.

385 27. Deslandes A. Comparative clinical pharmacokinetics of antibody-drug conjugates in first-in-

386 human phase 1 studies. MAbs 2014;6(4):859-70.

387 28. Hochhauser D, Meyer T, Spanswick VJ, Wu J, Clingen PH, Loadman P, et al. Phase I study

388 of sequence-selective minor groove DNA binding agent SJG-136 in patients with advanced solid

389 tumors. Clin Cancer Res 2009;15(6):2140-7.

390 29. Jain RK. Physiological barriers to delivery of monoclonal antibodies and other

391 macromolecules in tumors. Cancer Res 1990;50(3 Suppl):814s-9s.

392 30. Deonarain MP, Yahioglu G, Stamati I, Pomowski A, Clarke J, Edwards BM, et al. Small-

393 format drug conjugates: A viable alternative to ADCs for solid tumours? Antibodies 2018;7(2).

394 31. Proia DA, Smith DL, Zhang J, Jimenez JP, Sang J, Ogawa LS, et al. HSP90 inhibitor-SN-38

395 conjugate strategy for targeted delivery of topoisomerase I inhibitor to tumors. Mol Cancer Ther

396 2015;14(11):2422-32.

397 32. Bennett G, Lutz R, Park P, Harrison H, Lee K. Abstract 1167: Development of BT1718, a

398 novel bicycle drug conjugate for the treatment of lung cancer. Cancer Res 2017;77(13

399 Supplement):1167.

400 33. Eder M, Pavan S, Bauder-Wust U, van Rietschoten K, Baranski AC, Harrison H, et al.

401 Bicyclic peptides as a new modality for imaging and targeting of proteins overexpressed by

402 tumors. Cancer Res 2019;79(4):841-52.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

17

403 34. Li Z, Krippendorff BF, Sharma S, Walz AC, Lave T, Shah DK. Influence of molecular size on

404 tissue distribution of antibody fragments. MAbs 2016;8(1):113-9.

405 35. Thurber GM, Dane Wittrup K. A mechanistic compartmental model for total antibody uptake

406 in tumors. J Theor Biol 2012;314:57-68.

407 36. Gray R, Bradley R, Braybrooke J, Davies C, Pan H, Peto R, et al. Abstract GS1-01:

408 Increasing the dose density of adjuvant chemotherapy by shortening intervals between courses

409 or by sequential drug administration significantly reduces both disease recurrence and breast

410 cancer mortality: An EBCTCG meta-analysis of 21,000 women in 16 randomised trials. Cancer

411 Res 2018;78(4 Supplement):GS1-01.

412 37. Sparano JA, Wang M, Martino S, Jones V, Perez EA, Saphner T, et al. Weekly paclitaxel in

413 the adjuvant treatment of breast cancer. N Engl J Med 2008;358(16):1663-71.

414 38. Hinrichs MJM, Ryan PM, Zheng B, Afif-Rider S, Yu XQ, Gunsior M, et al. Fractionated

415 dosing improves preclinical therapeutic index of pyrrolobenzodiazepine-containing antibody

416 drug conjugates. Clin Cancer Res 2017;23(19):5858-68.

417 39. Day CP, Merlino G, Van Dyke T. Preclinical mouse cancer models: A maze of opportunities

418 and challenges. Cell 2015;163(1):39-53.

419 40. Tabrizi M, Funelas C, Suria H. Application of quantitative pharmacology in development of

420 therapeutic monoclonal antibodies. AAPS J 2010;12(4):592-601.

421 41. Gebhart G, Lamberts LE, Wimana Z, Garcia C, Emonts P, Ameye L, et al. Molecular

422 imaging as a tool to investigate heterogeneity of advanced HER2-positive breast cancer and to

423 predict patient outcome under trastuzumab emtansine (T-DM1): The ZEPHIR trial. Ann Oncol

424 2016;27(4):619-24.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

18

425 42. Massicano AVF, Marquez-Nostra BV, Lapi SE. Targeting HER2 in nuclear medicine for

426 imaging and therapy. Mol Imaging 2018;17:1536012117745386.

427 43. Herbertson RA, Tebbutt NC, Lee FT, MacFarlane DJ, Chappell B, Micallef N, et al. Phase I

428 biodistribution and pharmacokinetic study of lewis Y-targeting immunoconjugate CMD-193 in

429 patients with advanced epithelial cancers. Clin Cancer Res 2009;15(21):6709-15.

430 44. Scott AM, Tebbutt N, Lee FT, Cavicchiolo T, Liu Z, Gill S, et al. A phase I biodistribution and

431 pharmacokinetic trial of humanized monoclonal antibody Hu3s193 in patients with advanced

432 epithelial cancers that express the lewis-Y antigen. Clin Cancer Res 2007;13(11):3286-92.

433 45. Bergstralh DT, Sekelsky J. Interstrand crosslink repair: Can XPF-ERCC1 be let off the

434 hook? Trends Genet 2008;24(2):70-6.

435 46. Deans AJ, West SC. DNA interstrand crosslink repair and cancer. Nat Rev Cancer

436 2011;11(7):467-80.

437 47. Clauson C, Scharer OD, Niedernhofer L. Advances in understanding the complex

438 mechanisms of DNA interstrand cross-link repair. Cold Spring Harb Perspect Biol

439 2013;5(10):a012732.

440 48. Huang Y, Li L. DNA crosslinking damage and cancer - a tale of friend and foe. Transl

441 Cancer Res 2013;2(3):144-54.

442 49. Swisher EM, Lin KK, Oza AM, Scott CL, Giordano H, Sun J, et al. Rucaparib in relapsed,

443 platinum-sensitive high-grade ovarian carcinoma (ARIEL2 part 1): An international, multicentre,

444 open-label, phase 2 trial. Lancet Oncol 2017;18(1):75-87.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

19

445 50. Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, et al. Inhibition of poly(ADP-

446 ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 2009;361(2):123-34.

447 51. Fong PC, Yap TA, Boss DS, Carden CP, Mergui-Roelvink M, Gourley C, et al. Poly(ADP)-

448 ribose polymerase inhibition: Frequent durable responses in BRCA carrier ovarian cancer

449 correlating with platinum-free interval. J Clin Oncol 2010;28(15):2512-9.

450 52. Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Olaparib

451 maintenance therapy in platinum-sensitive relapsed ovarian cancer. N Engl J Med

452 2012;366(15):1382-92.

453 53. Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Olaparib

454 maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: A

455 preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial.

456 Lancet Oncol 2014;15(8):852-61.

457 54. Pilie P, Gay CM, Byers LA, O'Connor MJ, Yap TA. PARP inhibitors: Extending benefit

458 beyond BRCA mutant cancers. Clin Cancer Res 2019.

459 55. Coleman RL, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean A, et al. Rucaparib

460 maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy

461 (ARIEL3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet

462 2017;390(10106):1949-61.

463 56. Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, et al. Maintenance

464 olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med

465 2018;379(26):2495-505.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

20

466 57. Zhong H, Chen C, Tammali R, Breen S, Zhang J, Fazenbaker C, et al. Improved therapeutic

467 window in BRCA-mutant tumors with antibody-linked pyrrolobenzodiazepine dimers with and

468 without PARP inhibition. Mol Cancer Ther 2019;18(1):89-99.

469 58. Maede Y, Shimizu H, Fukushima T, Kogame T, Nakamura T, Miki T, et al. Differential and

470 common DNA repair pathways for topoisomerase I- and II-targeted drugs in a genetic DT40

471 repair cell screen panel. Mol Cancer Ther 2014;13(1):214-20.

472 59. Pommier Y. Topoisomerase I inhibitors: and beyond. Nat Rev Cancer

473 2006;6(10):789-802.

474 60. Zoppoli G, Regairaz M, Leo E, Reinhold WC, Varma S, Ballestrero A, et al. Putative

475 DNA/RNA helicase schlafen-11 (SLFN11) sensitizes cancer cells to DNA-damaging agents.

476 Proc Natl Acad Sci U S A 2012;109(37):15030-5.

477 61. Barrett A, Chen H, Howard P, Koopmann J, Kagiampakis I, Sridhar S. In: Analysis of large-

478 scale, high-throughput screens to identify genes which confer specific sensitivity to

479 pyrrolobenzodiazepine (PBD) warheads for ADCs. 3rd Exploring DNA Repair Pathways as

480 Targets for Cancer Therapy; Feb 23-26; 2019.

481 62. Odegaard JI, Vincent JJ, Mortimer S, Vowles JV, Ulrich BC, Banks KC, et al. Validation of a

482 plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-

483 based methodologies. Clin Cancer Res 2018;24(15):3539-49.

484 63. Rugo HS, Cortes J, Awada A, O'Shaughnessy J, Twelves C, Im SA, et al. Change in

485 topoisomerase 1-positive circulating tumor cells affects overall survival in patients with

486 advanced breast cancer after treatment with etirinotecan pegol. Clin Cancer Res

487 2018;24(14):3348-57.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

21

488 64. Fuh FK, Looney C, Li D, Poon KA, Dere RC, Danilenko DM, et al. Anti-CD22 and anti-

489 CD79b antibody-drug conjugates preferentially target proliferating B cells. Br J Pharmacol

490 2017;174(8):628-40.

491 65. Raja R, Kuziora M, Brohawn PZ, Higgs BW, Gupta A, Dennis PA, et al. Early reduction in

492 ctDNA predicts survival in patients with lung and bladder cancer treated with . Clin

493 Cancer Res 2018.

494 66. Matulonis UA, Moore KN, Martin LP, Vergote IB, Castro C, Gilbert L, et al. 949P

495 mirvetuximab soravtansine, a folate receptor alpha-targeting antibody-drug conjugate (ADC),

496 with pembrolizumab in platinum-resistant ovarian cancer (PROC): Initial results of an expansion

497 cohort from FORWARD II, a phase ib study. Annals of Oncology 2018;29(suppl_8):mdy285.157.

498 67. Emens L, Esteva F, Beresford M, Saura C, De Laurentiis M, Kim S, et al. Abstract PD3-01:

499 Results from KATE2, a randomized phase 2 study of atezolizumab (atezo)+trastuzumab

500 emtansine (T-DM1) vs placebo (pbo)+T-DM1 in previously treated HER2+ advanced breast

501 cancer (BC). Cancer Res 2019;79(4 Supplement):PD3-01.

502 68. Cao A, Heiser R, Law C, Gardai SJ. Abstract 4914: Auristatin-based antibody drug

503 conjugates activate multiple ER stress response pathways resulting in immunogenic cell death

504 and amplified T-cell responses. Cancer Res 2016;76(14 Supplement):4914.

505 69. Muller P, Martin K, Theurich S, Schreiner J, Savic S, Terszowski G, et al. Microtubule-

506 depolymerizing agents used in antibody-drug conjugates induce antitumor immunity by

507 stimulation of dendritic cells. Cancer Immunol Res 2014;2(8):741-55.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

22

508 70. Muller P, Kreuzaler M, Khan T, Thommen DS, Martin K, Glatz K, et al. Trastuzumab

509 emtansine (T-DM1) renders HER2+ breast cancer highly susceptible to CTLA-4/PD-1 blockade.

510 Science Translational Medicine 2015;7(315):315ra188.

511 71. Muller P, Rios-Doria J, Harper J, Cao A. Combining ADCs with immuno-oncology agents. in:

512 Innovations for next-generation antibody-drug conjugates. In: Humana Press; 2018. p. 11-44.

513 72. Rios-Doria J, Harper J, Rothstein R, Wetzel L, Chesebrough J, Marrero A, et al. Antibody-

514 drug conjugates bearing pyrrolobenzodiazepine or tubulysin payloads are immunomodulatory

515 and synergize with multiple immunotherapies. Cancer Res 2017;77(10):2686-98.

516 73. Diem S, Hasan Ali O, Ackermann CJ, Bomze D, Koelzer VH, Jochum W, et al. Tumor

517 infiltrating lymphocytes in lymph node metastases of stage III correspond to

518 response and survival in nine patients treated with at the time of stage IV disease.

519 Cancer Immunol Immunother 2018;67(1):39-45.

520 74. Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab

521 versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med

522 2016;375(19):1823-33.

523 75. Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab

524 versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer

525 (KEYNOTE-010): A randomised controlled trial. Lancet 2016;387(10027):1540-50.

526 76. Higgs BW, Morehouse CA, Streicher K, Brohawn PZ, Pilataxi F, Gupta A, et al. Interferon

527 gamma messenger RNA signature in tumor biopsies predicts outcomes in patients with non-

528 small cell lung carcinoma or urothelial cancer treated with durvalumab. Clin Cancer Res

529 2018;24(16):3857-66.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

23

530 77. Bardia A, Vahdat L, Diamond J, Kalinsky K, O'Shaughnessy J, Moroose R, et al. In:

531 Sacituzumab govitecan (IMMU-132), an anti-trop-2-SN-38 antibody-drug conjugate, as ³3rd-line

532 therapeutic option for patients with relapsed/refractory metastatic triple-negative breast cancer

533 (mTNBC): Efficacy results. Dec 5-9, 2017; SABCS Abstract Book: San Antonio Breast Cancer

534 Symposium; 2017.

535 78. Bardia A, Robinson Diamond J, Vahdat L, M. Tolaney S, O'Shaughnessy J, Moroose R, et

536 al. Efficacy of sacituzumab govitecan (anti-trop-2-SN-38 antibody-drug conjugate) for treatment-

537 refractory hormone-receptor positive (HR+)/HER2- metastatic breast cancer (mBC). Journal of

538 Clinical Oncology 2018;36:1004.

539 79. Tagawa ST, Faltas B, Lam E, Saylor P, Bardia A, Hajdenberg J, et al. 858P Sacituzumab

540 govitecan (IMMU-132) for patients with pretreated metastatic urothelial uancer (UC): Interim

541 results. Annals of Oncology 2017;28(suppl_5):md371.012.

542 80. Iwata H, Tamura K, Doi T, Tsurutani J, Modi S, Park H, et al. Trastuzumab deruxtecan (DS-

543 8201a) in subjects with HER2-expressing solid tumors: Long-term results of a large phase 1

544 study with multiple expansion cohorts. Journal of Clinical Oncology 2018;36(15\_suppl):2501.

545 81. Kogawa T, Yonemori K, Masuda N, Takahashi S, Takahashi M, Iwase H, et al. Single agent

546 activity of U3-1402, a HER3-targeting antibody-drug conjugate, in breast cancer patients: Phase

547 1 dose escalation study. Journal of Clinical Oncology 2018;36(15\_suppl):2512.

548 549 550

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

24

Figures and Tables Table 1. Topoisomerase I–targeted warheads demonstrate robust clinical efficacy

ADC Target/warhead Population ORR (%) DCR (%) DOR PFS (months) (months)

Sacituzumab TROP-2/SN-38 ≥ 3L TNBC 31 (6 CRs) 46 7.6 5.5 govitecan (77- 79) irinotecan metabolite (n = 110) + Immunomedics (topoisomerase) ≥ 2L HR BC 31 (0 CRs) 63 7.4 6.8 (n = 54)

≥ 2L UC 34 (2 CRs) 49 13 7.1

(n = 41)

Trastuzumab HER2/exetecan ≥ 3L HER2-high BC (n = 55 94 Not reached Not reached deruxtecan (80) 111) (DS-8201a) ≥ 2L HER2-low BC 50 85 11 13

Daiichi Sankyo (n = 34)

≥ 3L HER2+ gastric 43 80 7.0 5.6

(n = 44)

≥ 3L HER2+ others 39 84 13 12

(n = 51; CRC, NSCLC +)

U3-1402 (81) HER3/exetecan ≥ 3L HER3+ BC 47 94 Not reported Not reported

Daiichi Sankyo (n = 32)

3L, third line; ADC, antibody drug conjugate; BC, breast cancer; CR, complete response; CRC, colorectal cancer; DCR, disease control rate; DOR, duration of response; HR, hormone receptor; NSCLC, non-small cell lung cancer; ORR, objective response rate; PFS, progression-free survival; TNBC, triple-negative breast cancer; UC, urothelial cancer.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

25

FIGURE LEGENDS Figure 1. ADC structure and therapeutic index optimization strategies. ADCs comprised a tumor-specific antibody, a linker, and a cytotoxic payload. Advances in chemistry of all 3 components are underway to potentially increase the therapeutic index. CDR, complement- determining region; DAR, drug-antibody ratio; MOA, mechanism of action.

Figure 2. ADCs in clinical development. A, ADCs in clinical development as of March 2019 shown by phase of development, indication and warhead according to clinicaltrials.gov. B, ADCs in combination with checkpoint inhibitors in clinical development, shown by phase of development, indication and warhead employed. Mylotarg and Besponsa are manufactured by Pfizer; Adcetris is manufactured by Seattle Genetics, Inc.; Kadcyla is manufactured by Genentech. Atezo, atezolizumab; Durva, durvalumab; Ipi, ipilimumab; Lonca-T, ; Mirve-S, mirvetuximab soravtansine; Nivo, ; Pembro, pembrolizumab; Pola- V, polatuzumab vedotin; Rova-T, rovalpituzumab terisine; Saci-G, sacituzumab govitecan; Teliso-V, telisotuzumab vedotin; Tiso-V, tisotumab vedotin.

Figure 3. Translational medicine strategies to maximize the therapeutic index. One of the key challenges for the clinical development of ADCs is the narrow index observed between safety and efficacy. The design and application of biomarkers to optimize patient selection, capture response signals early, and inform potential combination therapies is central to maximizing the therapeutic index and providing a personalized approach to ADC therapeutic development.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 1:

Conjugation/linker đ Several chemistries đ Multiple DARs đ Tumor-specific triggers

Antibody đ Formats: i.e., Ab fragments đ Half-life extension đ CDR-masking technologies đ Enhance drug delivery Payload đ Match right payload MOA for right target/patient population đ Alternative warheads: i.e., targeted agents in both tumor and tumor microenvironment

© 2019 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2:

A PHASE Acute er* mye th leuke loi O mia d s or ) I Mu um ed my lti t ifi el pl id ec om e ol p S ns a (u B -c e n ll ll Tiso-V II o m l n e y - a c Anetumab GSK2857916 m H li l p o g a ravtansine h d n o g a n m n Indatuximab k c e i a n ie R ravtansine s III Lonca-T / Pola-V C AGS-16C3F Naratuximab o r l e M emtansine o r d Mylotarg K e d c

a t l T a

B Trastuzumab Enfortumab Adcetris Trastuzumab l deruxtecan vedotin Besponsa deruxtecan

H

s

e q

a u

d a c

m i a Trastuzumab r n o t d u deruxtecan s s n a Kadcyla c e G e c l k l Trastuzumab Mirve-S deruxtecan Rova-T

P Trastuzumab c r i o Saci-G t s duocarmazine a t Tiso-V re a Teliso-V c te n a SAR566658 Trastuzumab P RC48 deruxtecan g O Mirve-S n v lu ar ll ia ce n all Sm HE ng R2 ll lu + b ll ce reast -sma Non-HER2+ breast Non

*Includes neuroendocrine, esophageal, glioblastoma multiforme, cervical, mesothelioma, and melanoma tumors. Microtubule inhibitor Topoisomerase inhibitor DNA damaging Mechanism unknown

B PHASE HE ME er dd I la Lonca-T + B Durva Trastuzumab deruxtecan + Nivo Pola-V Saci-G + Durva + Atezo II S Brentuximab vedotin + Teliso-V + Nivo o Nivo and/or lpi Anetumab ravtansine + li + GSK2857916 + Pembro lpi + Nivo d t Pembro PF-066447020 + u Tiso-V + Pembro m III Brentuximab vedotin + Nivo o Brentuximab r vedotin + Pembro Rova-T + ABBV-181 s BMS-986148 + M SC006 + ABBV-181 n Nivo Mirve-S + MGC018 + MGA012 a K i Pembro r T a BMS-986148 + Nivo SC004 + ABBV-181 v

O

Mirve-S + BMS-986148 + a Pembro Nivo m o i l

e

Anetumab h t ravtansine o + Pembro s Trastuzumab emtansine e + Pembro M Trastuzumab Ladiratuzumab vedotin Anetumab ravtansine + Atezo emtansine + + Pembro Teliso-V + Nivo Atezo

Trastuzumab emtansine Trastuzumab + utomilumab deruxtecan + Pembro Trastuzumab deruxtecan + Nivo B BMS-986148 + re Trastuzumab deruxtecan + Pembro Nivo as Saci-G + Durva Rova-T + Nivo +/– lpi t SC011 + ABBV-181 g Lun

Microtubule inhibitor Mechanism unknown DNA damaging Checkpoint inhibitor Topoisomerase inhibitor Co-stimulation agonist

© 2019 American Association for Cancer Research Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 3:

Optimize patient Develop blood biomarkers to capture selection response signals early (ctDNA)

đ Mechanism-based biomarkers for đ Surrogate markers of tumor burden enrichment and selection of patients đ Mutational profiles of response

Leverage experience to inform combinations

đ Target immunologically responsive tumor types đ Evaluate biological responses post ADC that predict response to IO

© 2019 American Association for Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Antibody Drug Conjugates: Future Directions in Clinical and Translational Strategies to Improve the Therapeutic Index

Steven Coats, Marna Williams, Benjamin Kebble, et al.

Clin Cancer Res Published OnlineFirst April 12, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-19-0272

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2019/04/12/1078-0432.CCR-19-0272. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2019 American Association for Cancer Research.