UB-311, a novel UBITh amyloid-β peptide

0 downloads 0 Views 1MB Size Report
Apr 14, 2017 - Chang Yi Wang a,b,c,*, Pei-Ning Wang d ...... Vaccine 2002;21:89–97. [29] Wang CY, Walfield AM, Fang XD, Hammerberg B, Ye J, Li ML, et al.
Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56

Featured Article Q1

Q9

UB-311, a novel UBITh amyloid-b peptide vaccine for mild Alzheimer’s disease Chang Yi Wanga,b,c,*, Pei-Ning Wangd, Ming-Jang Chiue, Connie L. Finstada, Feng Lina, Shugene Lynnb, Yuan-Hung Taib, Xin De Fanga, Kesheng Zhaoa, Chung-Ho Hunga, Yiting Tsengc, Wen-Jiun Pengb, Jason Wangb, Chih-Chieh Yuc, Be-Sheng Kuoa, Paul A. Frohnac a United Biomedical, Inc. (UBI), Hauppauge, NY, USA United Biomedical Inc., Asia (UBI-Asia), Hsinchu, Taiwan c United Neuroscience, Inc. (UNS), Hsinchu, Taiwan d Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan e Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan

Q2

b

Abstract

Introduction: A novel amyloid-b (Ab) synthetic peptide vaccine (UB-311) has been evaluated in a first-in-human trial with patients of mild-to-moderate Alzheimer’s disease. We describe translational research covering vaccine design, preclinical characterization, and phase-I clinical trial with supportive outcome that advances UB-311 into an ongoing phase-II trial. Methods: UB-311 is constructed with two synthetic Ab1–14–targeting peptides (B-cell epitope), each linked to different helper T-cell peptide epitopes (UBITh) and formulated in a Th2-biased delivery system. The hAPP751 transgenic mouse model was used to perform the proof-of-concept study. Baboons and macaques were used for preclinical safety, tolerability, and immunogenicity evaluation. Patients with mild-to-moderate Alzheimer’s disease (AD) were immunized by intramuscular route with 3 doses of UB-311 at weeks 0, 4, and 12, and monitored until week 48. Safety and immunogenicity were assessed per protocol, and preliminary efficacy was analyzed by Alzheimer’s Disease Assessment Scale–Cognitive Subscale (ADAS-Cog), Mini–Mental State Examination (MMSE), and Alzheimer’s Disease Cooperative Study–Clinician’s Global Impression of Change. Results: UB-311 covers a diverse genetic background and facilitates strong immune response with high responder rate. UB-311 reduced the levels of Ab1–42 oligomers, protofibrils, and plaque load in hAPP751 transgenic mice. Safe and well-tolerated UB-311 generated considerable site-specific (Ab1–10) antibodies across all animal species examined. In AD patients, UB-311 induced a 100% responder rate; injection site swelling and agitation were the most common adverse events (4/19 each). A slower rate of increase in ADAS-Cog from baseline to week 48 was observed in the subgroup of mild AD patients (MMSE  20) compared with the moderate AD subgroup, suggesting that UB311 may have a potential of cognition improvement in patients with early stage of Alzheimer’s dementia. Discussion: The UBITh platform can generate a high-precision molecular vaccine with high responder rate, strong on-target immunogenicity, and a potential of cognition improvement, which support UB-311 for active immunotherapy in early-to-mild AD patients currently enrolled in a phase-II trial (NCT02551809). Ó 2017 Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Keywords:

UB-311; UBITh platform; Amyloid-b vaccine; FIH clinical trial; Alzheimer’s disease

*Corresponding author. Tel.: 631-273-2828; Fax: 631-273-1717. E-mail address: [email protected] http://dx.doi.org/10.1016/j.trci.2017.03.005 2352-8737/Ó 2017 Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an open access article under the CC BY-NC-ND license (http:// creativecommons.org/licenses/by-nc-nd/4.0/). FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117

2

118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

1. Introduction The amyloid beta (Ab) peptide, central to the “Amyloid Cascade Hypothesis,” is thought to be the pivot for the onset and progression of Alzheimer’s disease (AD), and the toxic forms of oligomers and fibrils are suggested to be responsible for the death of synapses and neurons that leads the pathology of AD and dementia [1,2]. Although recent findings may suggest a dual protective/damaging role for Ab peptides in AD pathology [3,4], a successful disease-modifying therapy for AD will include products that affect the disposition of Ab in the brain [5], in which the immunotherapeutic strategy has drawn much attention since the first active immunotherapy was investigated in mice in 1999 [6]. AN-1792 vaccine [7,8], using aggregated full-length Ab1–42 peptide as immunogen associated with a Th1 adjuvant (QS-21; saponin), was the first immunotherapy tested in AD patients, which generated anti-Ab antibody responses in ,25% of patients with improved memory and decreased level of tau protein in the cerebral spinal fluid of a small subset of patients [7,8], thus providing encouraging results for development of new Ab vaccines. AN-1792 was discontinued because of acute meningoencephalitis symptoms in 6% of patients, likely caused by autoreactive T-cell activation and Ab-reactive T-cell infiltration into the central nervous system [7]. Several second-generation Ab-targeting vaccines were subsequently designed to minimize Ab-related T-cell inflammation. These include the following: ACC-001 using Ab1–7 peptide conjugated to diphtheria toxoid protein [9], CAD106 using Ab1–6 peptide coupled to Qb virus-like particle [10], V950 using multivalent Ab1–15 conjugated to a carrier unknown [11], and affitopes AD01 and AD02 using Ab mimetics (six amino acids) conjugated to KLH [12]. These vaccines induced variable anti-Ab antibody titers and responder rates, with most of the immune response directed toward the large carrier molecules. To date, these and related vaccines have not presented convincing clinical efficacy data [13]. Development of AD02, ACC-001, and V950 vaccines have been discontinued for reasons unclear [14]. CAD106, currently in clinical phase-III trial, has completed two phase-II trials reporting acceptable safety and tolerability and evoking a strong serological responses in 80% of patients, and brain PET imaging was suggestive of target engagement [15,16]. Amyloid-b is the principal target of late-stage development programs with relatively few agents in clinical trials for AD, suggesting a need to amplify the drug discovery ecosystem [17]. We describe in this report a novel design of the UBITh platform-based, fully synthetic Ab1–14 peptide vaccine (UB-311), preclinical characterization, and a first-inhuman (FIH) phase-I clinical study which enrolled 19 patients with mild-to-moderate AD. UB-311 comprises two Ab1–14–targeting peptides (B-cell epitopes), each linked to different helper T-cell peptide epitopes (UBITh) as a chimeric peptide to maximize immunogenicity, which is

formulated in a Th2-biased delivery system to minimize Tcell inflammatory reactivity. The UB-311 vaccine was safe and well-tolerated, generating strong site-specific (Ab1–10) antibodies in all patients. Of note, a subgroup of mild AD patients (Mini–Mental State Examination [MMSE] 20) showed a positive trend toward cognition improvement. 2. Methods 2.1. Peptide synthesis and vaccine product UBITh Ab1–14 peptide immunogens for the UB-311 vac- Q3 cine product and Ab peptide antigens for immunoassays were synthesized using automated solid-phase synthesis, purified by preparative HPLC, and characterized by MALDI-TOF mass spectrometer, amino acid analysis, and reverse-phase HPLC. UB-311 comprises two Ab1–14–targeting peptides (B-cell epitope), each synthetically linked to different helper T-cell peptide epitopes (UBITh), and formulated in an alum-containing Th2-biased delivery system (US patent no. 9,102,752). The two Ab immunogens are the cationic Ab1–14-εK-KKK-MvF5 Th [ISITEIKGVIVHRIETILF] and Ab1–14-εK-HBsAg3 Th [KKKIITITRIITIITID] peptides, in equimolar ratio; they were mixed with polyanionic CpG oligodeoxynucleotide (ODN) to form stable immunostimulatory complexes of micron-size particulates, to which aluminum mineral salt (Adju-Phos, Brenntag Biosector, Denmark), was added to the final formulation, along with sodium chloride for tonicity and 0.25% 2phenoxyethanol as preservative. The vaccine product was Q4 manufactured under GMP conditions at United Biomedical as sponsor (New York). 2.2. FIH phase-I clinical trial with patients of mild-tomoderate Alzheimer’s disease 2.2.1. Patients and study design An FIH phase-I clinical trial (NCT00965588) with a 24week intervention phase (Fig. 1) was conducted at National Taiwan University Hospital and Taipei Veterans General Hospital in Taipei, Taiwan. A total of 19 adults with mildto-moderate AD were enrolled, including 9 males and 11 females with 50–85 years of age (Table 1). Each patient was immunized three times with UB-311 (300 mg/dose) at weeks 0 (baseline), 4, and 12 by intramuscular injection. An observational 24-week extension study (NCT01189084), from weeks 24 to 48, was included to monitor long-term immunogenicity and efficacy with 14 eligible subjects (Fig. 1). 2.2.2. Objectives of the FIH trial The primary objectives were to evaluate safety and tolerability; the secondary objectives were to assess immunogenicity and the preliminary vaccine efficacy after a three-dose regimen. Safety assessments included vital signs, physical and neurological examinations, laboratory parameters, electrocardiogram,

FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300

3

Fig. 1. Patient disposition and study design. The first-in-human (FIH) clinical trial enrolled 19 patients (50–80 years old) with mild-to-moderate Alzheimer’s disease in a 24-week interventional study, and all 19 subjects received three UB-311 vaccine doses by intramuscular injection (300-mg immunogen peptide) at 0, 4, and 12 weeks and completed the treatment study (identifier no.: NCT00965588). After the first five subjects passed the safety evaluation at week 48, the remaining 14 subjects enrolled in and completed a 24-week observational extension study added to the end of the interventional study to monitor long-term safety, tolerability, immunogenicity, and efficacy (identifier no.: NCT01189084).

Table 1 Baseline demographics and characteristics of patients with Alzheimer’s disease (first-in-human trial with UB-311 vaccine) Baseline demographics and disease assessment* Age, years Mean (SD) Median (range) Gender Male (%) Female (%) Race Asian (%) Other (%) BMI, kg/m2 Mean (SD) Median (range) Time since Alzheimer’s disease diagnosis, years Mean (SD) Median (range) ADCS-CGIC Borderline (%) Mild (%) Moderate (%) MMSE Mean (SD) Median (range) HIS Mean (SD) Median (range) CDR Mean (SD) Median (range)

Value 64.0 (8.3) 64.0 (51.0–78.0) 9 (47.4) 10 (52.6) 19 (100.0) 0 (0.0) 23.1 (2.1) 22.6 (20.0–27.2)

2.4 (2.3) 2.0 (0.3–10.7) 5 (26.3) 11 (57.9) 3 (15.8) 19.2 (3.2) 19.0 (15.0–25.0) 1.4 (1.2) 1.0 (0.0–4.0) 1.1 (0.3) 1.0 (1.0–2.0)

Abbreviations: SD, standard deviation; BMI, body mass index; ADCSCGIC, Alzheimer’s Disease Cooperative Study–Clinical Global Impression of Change; MMSE, Mini–Mental State Examination; HIS, Hachinski Ischemic Score; CDR, Clinical Dementia Rating. *The enrolled patients were free of psychiatric, medical or substance abuse problems, history of severe systemic, autoimmune disease or anaphylaxis, hepatic insufficiency, poor control of blood sugar, positive human immunodeficiency virus, hepatitis C virus antibodies, or hepatitis B surface antigen enzyme immunoassay test results. Patients receiving stable treatment with cholinesterase inhibitors were not excluded from the trial.

and brain magnetic resonance imaging (MRI) scans were performed at the initial screening and at the end of the interventional study (weeks 24–26). No abnormal MRI brain imaging scans were reported. Blood samples were collected periodically for clinical laboratory analysis, screening, and characterization of anti-Ab antibodies, and for in vitro lymphocyte proliferation and cytokine production. 2.2.3. Preliminary evaluation of efficacy The effect of the vaccine on cognition responses was evaluated using three scales: two cognitive scales, Alzheimer’s Disease Assessment Scale–Cognitive Subscale (ADAS-Cog) and MMSE; and a global rating scale, Alzheimer’s Disease Cooperative Study–Clinician’s Global Impression of Change (ADCS-CGIC). ADAS-Cog and ADCS-CGIC were performed at weeks 0, 16, 24, and 48; MMSE was performed at screening visit, and at weeks 16, 24, 36, and 48. 2.2.4. Statistical analyses For the UB-311 FIH trial, 19 treated patients were recruited to demonstrate initial safety; the probability of observing at least one case of meningoencephalitis with a true incidence of 6% was calculated to be 69.1%. Pearson’s correlation coefficient was calculated for the relationship assessment between anti-Ab antibody titer and ADAS-Cog and MMSE change. For potential heterogeneity in treatment effects, subgroup analyses were conducted on the efficacy end points by baseline age and MMSE. Repeatedmeasures mixed-effects model was used to estimate the rates of change (slope) in ADAS-Cog and MMSE scores and to determine the difference in slope between different subgroups. The model terms included baseline efficacy variable, subgroup variable, visit time, and the interaction between subgroup variable and visit time. First-order autocorrelation was used as the covariance matrix based on the Akaike’s information criterion. All statistical analyses were based on two-sided tests at a significance level of 0.05 and conducted

FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361

4

with R version 2.14.1 (R Foundation for Statistical Computing, Vienna, Austria).

3. Results

2.3. Supplementary methods online

UB-311 comprises two peptide immunogens, each with an N-terminal Ab1–14 peptide, synthetically linked through an amino acid spacer to different T helper (Th) cell peptides (UBITh epitopes) derived from two pathogen proteins, hepatitis B surface antigen (Supplementary Fig. 1A), and measles virus fusion protein (Supplementary Fig. 1B). The vaccine product, formulated in CpG ODN and alumcontaining Th2-biased delivery system, could maintain long-term chemical stability as demonstrated after storage for 3 years at 2 C–8 C (Supplementary Fig. 1C). Each peptide was purified to a well-defined molecular entity with an exact mass close to its respective theoretical value (Supplementary Figs. 1D and 1E). The unique design with UBITh peptide epitopes (MvF5 and HBsAg3) allows for maximal coverage of the major

Supplementary methods and study results are provided online to support specific preclinical studies in animals and additional clinical data: (1) experimental animals for immunogenicity, pharmacology, and toxicity studies; (2) hAPP751 transgenic mouse model study; (3) enzymelinked immunosorbent assay (ELISA) test for detection of antibodies to synthetic peptides; (4) specificity analyses of anti-Ab antibodies by epitope mapping solid-phase ELISA for detection of b-amyloid antigens; (6) immunohistochemical analysis of human tissue cross-reactivity; (7) Biochemical extraction of amyloid-b from hAPP751 mouse brain tissues; (8) lymphocyte proliferation and cytokine analyses; and (9) dot-blot analysis of Ab forms reactive with human antisera.

3.1. High levels of anti-Ab antibody response and responder rate

web 4C=FPO

362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

Fig. 2. Serum anti-Ab antibody titers assayed by ELISA and preferable targeted Ab species visualized by dot plot after UB-311 immunization in AD patients. (A) Mean antibody response during the 24-week interventional study (solid line) in 19 AD patients treated with UB-311; 14 of 19 patients (dashed line) were followed up in an additional 24-week observational study, whose mean ADAS-Cog score was 4.9 (improvement). The mean baseline titer (pretreatment) was 1.0 log10; and 4 weeks after last vaccine boost at week 16, the titer peaked at 2.7 log10 (range: 1.8–3.7 log10). At week 48, all patients had decreasing but still positive antibody titers, measured by Ab1–28 ELISA test. At week 16, serum samples recognized Ab1–42 monomers (B) and oligomers (C); preimmune serum samples collected at week 0 had anti-Ab1–42 antibody levels below quantification limit (not shown on log scale), except subjects P109 (monomer) and P108 (oligomer). (D) At week 16, analysis of serum dot plot (left panel), flanked by the positive control 6E10 mAb, reveals that the vaccine-induced anti-Ab antibodies bind preferentially to Ab fibrils, followed by oligomers, and the least to monomers; the densitometric measures (right panel) for the three Ab species are the mean scales from three representative AD subjects, P105, P108, and P206. Abbreviations: AD, Alzheimer’s disease; ADAS-Cog, Alzheimer’s Disease Assessment Scale–Cognitive Subscale; ELISA, enzyme-linked immunosorbent assay. FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544

5

Table 2 Site specificity of anti-Ab antibody in patients by peptide epitope mapping Peptide sequence TEEISEVKMD EEISEVKMDA EISEVKMDAE ISEVKMDAEF SEVKMDAEFR EVKMDAEFRH VKMDAEFRHD KMDAEFRHDS MDAEFRHDSG DAEFRHDSGY AEFRHDSGYE EFRHDSGYEV FRHDSGYEVH RHDSGYEVHH HDSGYEVHHQ DSGYEVHHQK SGYEVHHQKL GYEVHHQKLV YEVHHQKLVF EVHHQKLVFF VHHQKLVFFA HHQKLVFFAE HQKLVFFAED QKLVFFAEDV DAEFRHD SGYEVHHQ KLVFFAED VGSNK

Subjects Ab position P101 P102 P103 P104 P105 P106 P107 P108 P109 P201 P202 29 to 1 0 0 0 28 to 2 0 0 0 27 to 3 0 0 0 26 to 4 0 0 0 25 to 5 10 0 0 24 to 6 1 0 0 23 to 7 31 0 0 22 to 8 94 0 0 21 to 9 420 12 0 1–10 1983 1669 36 2–11 2 0 0 3–12 0 0 0 4–13 0 0 0 5–14 0 0 0 6–15 0 0 0 7–16 214 0 0 8–17 0 0 0 9–18 0 0 0 10–19 0 0 0 11–20 0 0 0 12–21 0 0 0 13–22 0 0 0 14–23 0 0 0 15–24 0 0 0 1–28 685 161 220

0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1721 3840 5193 2838 948 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 295 450 715 1163 478

P203 P204 P205 P206 P207 P208 P209

0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 58 0 0 0 0 0 0 0 1 0 2 7 0 12 0 19 116 0 17 1675 1034 42,313 1284 274 0 3 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 430 339 509 279 284

0 0 0 0 0 0 0 0 0 81 0 0 0 0 0 0 0 0 0 0 0 0 0 0 39

P210

0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 1 0 0 0 0 0 0 0 13 0 0 0 0 52 0 2 6 0 121 1797 6024 4093 27,596 20,968 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 16 0 0 0 0 10 0 0 0 0 0 0 0 0 0 27 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 226 906 1157 893 1321

NOTE. The in vitro anti-Ab antibody epitope mapping is a competitive binding inhibition enzyme-linked immunoassay for determination of binding residues Q8 of anti-Ab antibody within the target region; the method is described in Section 2. The N-terminal Ab1–10 (DAEFRHDSGY) peptide (given in bold) was identified to react most strongly with the serum samples collected on week 16 from each of the 19 patients immunized with UB-311; the results show 100% responder rate after immunization at weeks 0, 4, and 12. The numbers represent the dilution factor of each serum sample that corresponds to the 50% binding inhibition (IC50) by each of the 24 Ab 10-mer peptides.

histocompatibility complex class II binding motifs that can facilitate a high responder rate among the patient population with diverse genetic backgrounds (Supplementary Fig. 2). Of interest, in B-cell antigenicity, UB-311 elicited a strong antibody response that was exclusively directed to the vaccine target Ab1–14 but not to the UBITh epitopes, as shown representatively in guinea pigs (Supplementary Table 1). UB-311 can elicit rapid recall immune responses as shown in vaccinated baboons after a long rest period of 72 weeks [18]. UB-311 generated high immune responses with 100% responder rate in diverse animal species (Supplementary Table 2) and durable, high anti-Ab antibody response (Fig. 2A) in all of 19 AD patients in the FIH trial (Figs. 2B and 2C; Table 2). UB-311 generated, over-thethreshold, a mean anti-Ab antibody titer of 1.7 log10 at week 8 after two immunizations (weeks 0 and 4); the immune response peaked with 2.7 log10 at week 16 (after booster injection at week 12) and remained elevated at week 48 (Fig. 2A).

3.2. Site-specificity epitope mapping The UBITh platform can generate a high-precision molecular vaccine as UB-311 has been shown, with immunized sera from baboons [18], to bind a precise peptide sequence DAEFRHDSGY, which represents the N-terminal 10-mer peptide of the full-length Ab1–42. In an additional competitive ELISA (Supplementary Table 3), significant reactions were seen with Ab1–28 and Ab1–10 peptides only (equivalent high inhibition level), but not with adjoining residue masking “D” (Ab[21]–9) or N-terminal “D” removed (Ab2–11, Ab3–12) or nonimmunogen peptide Ab17–43. The exactness of binding-site specificity at DAEFRHDSGY was further confirmed by 10-mer epitope mapping using sera from all 19 patients with AD (Table 2).

3.3. Preferential Ab forms targeted by the vaccine At week 16 after vaccination in AD patients where peak anti-Ab responses were observed (Fig. 2A), antisera on

FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605

6

606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

ELISA was found to recognize Ab1–42 monomers (Fig. 2B) and oligomers (Fig. 2C). Further analysis with dot blot (Fig. 2D) reveals that the vaccine-induced, anti-Ab IgGs (purified from antisera) bind preferentially to Ab fibrils, followed by oligomers, and the least to monomers. 3.4. Safety features in animals and human subjects In UB-311-treated hAPP751 transgenic mice and cynomolgus macaques, no evidence was found for microglial cell activation (anti-CD11b antibody marker) or for T-cell infiltration (anti-CD3 antibody marker) of brain tissue; no changes in lymphocyte proliferation or upregulation of cytokine secretion were noted in macaque peripheral blood mononuclear cells (PBMCs) incubated with Ab1–14 peptide [18]. In the FDA-approved panel of 30 normal adult human tissues stained with purified guinea pig anti-Ab1–14 IgG, all tissues were negative other than some positive beta-amyloid plaques and cerebral vessels in some brain samples from

elderly individuals (Supplementary Fig. 3), indicative of no nonspecific binding by immunoglobulins generated after UB-311 vaccination. UB-311 was safe and well tolerated in the 19 AD patients of FIH trial. Injection site swelling (4/19, 21%), agitation (4/ 19, 21%), and transient increase in alanine aminotransferase levels in blood (3/19, 16%) were the most common adverse effects. Treatment-emergent adverse events in 19 AD patients were reported in Table 3. No significant changes were notable with the electrocardiograms, brain magnetic resonance imaging scans, and neurology examination. Laboratory measures of hematological tests, blood clinical chemistry tests, and coagulation factors showed stable trends throughout the study period. No change of lymphocyte proliferation responses (Supplementary Table 4) or cytokine upregulation (Supplementary Table 5) was observed in the presence of Ab1–14 or Ab1–28 peptides. Of special note, no evidence of meningoencephalitis was recorded during or after the study.

Table 3 Phase-I treatment-emergent adverse events in 19 patients with AD Adverse event by preferred term System organ class*

Preferred term

No. of subjects 5 19

Gastrointestinal disorders

Toothache Vomiting Injection site hemorrhage Injection site pain Injection site swelling Herpes zoster Upper respiratory tract infection Burns first degree Open wound Alanine aminotransferase increased Aspartate aminotransferase increased Autoantibody positive Neutrophil count increased Neutrophil percentage increased Red blood cell sedimentation rate increased White blood cell count increased Decreased appetite Hyperglycemia Poor quality sleep Dizziness Agitation Obsessive-compulsive disorder Genitourinary tract infection Nephrolithiasis Epididymitis Cough Herpes zoster Cellulitis Tinea cruris

1 (5.3%) 1 (5.3%) 1 (5.3%) 1 (5.3%) 4 (21.1%) 1 (5.3%) 2 (10.5%) 1 (5.3%) 1 (5.3%) 3 (15.8%) 2 (10.5%) 2 (10.5%) 1 (5.3%) 1 (5.3%) 2 (10.5%) 1 (5.3%) 1 (5.3%) 1 (5.3%) 1 (5.3%) 1 (5.3%) 4 (21.1%) 1 (5.3%) 1 (5.3%) 1 (5.3%) 1 (5.3%) 2 (10.5%) 1 (5.3%) 1 (5.3%) 1 (5.3%)

General disorders and administration site conditions

Infections and infestations Injury, poisoning, and procedural complications Investigationsy

Metabolism and nutrition disorders Nervous system disorders Psychiatric disorders Renal and urinary disorders Reproductive system and breast disorders Respiratory, thoracic and mediastinal disorders Skin and subcutaneous tissue disorders

Abbreviation: AD, Alzheimer’s disease. NOTE. Summary of treatment-emergent adverse events (AEs). The MedDRA system was used to code the AEs. *The AEs were summarized descriptively by system organ class and preferred terms. AEs were classified into pretreatment and treatment-emergent AEs according to the time of occurrence. Treatment-emergent AEs which were definitely, probably, or possibly related to the test drugs were regarded as treatment-related AEs. There were a total of 16 treatment-related AEs in 9 (47.4%) subjects. y Laboratory evaluation.

FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

3.5. Functional immunogenicity in animals and human subjects UB-311 reduced plaque size in the cortex and hippocampus of hAPP751 aged responder transgenic mice (Supplementary Q5 Fig. 4A). The reducing effects correlated with the lowering of Ab1–42 levels in brain tissue extracts, in which young responder transgenic mice were found to have fewer protofibrils (Triton X-100 fraction) and fewer large oligomers and fibrils (SDS fraction) compared with nontreated or placebo control animals (Supplementary Fig. 4B). In cynomolgus macaques, UB-311 promoted brain-toblood Ab efflux as demonstrated with increased plasma Ab1–40 levels in the presence of elevated anti-Ab1–14 antibody levels (Supplementary Table 6); Ab1–40 levels remained steady state in the cerebral spinal fluid [18]. This was also observed in AD patients, although anti-Ab1–14 antibody titers were much higher in the macaques after six vaccine doses than those in human subjects after three immunizations. Serum Ab1–40 levels from 9 of 12 patients were measured for net change; anti-Ab1–14 antibody titers greater than 2.4 log10 in serum showed measurable efflux (Supplementary Table 7). Of note, serum antibodies from all AD patients could bind Ab1–42 monomers and oligomers (Figs. 2B and 2C), an activity that may lead to reduction of soluble oligomers in the brain.

3.6. Potential efficacy for mild Alzheimer’s disease On a 3-dose UB-311 vaccine regimen in FIH trial Q6 (300 mg/dose UB-311 at weeks 0, 4, and 12) and monitored through week 24 (19 patients in interventional study phase) and week 48 (14 patients in observational extension study phase) (Fig. 1). Observed in the 14 patients, ADAS-Cog scores increased by 3.2 points, MMSE scores decreased by 2.2 points, and about 39% of patients showed improvement or no change in ADCS-CGIC scores derived based on the clinician’s perception of the patient’s change in global clinical status over time. Possible confounder effects in the subgroups based on the age component, baseline MMSE scores, or gender were further analyzed in the post hoc subgroup analyses (Fig. 3). A significantly lower rate of change in ADAS-Cog from baseline to week 48 was observed in the subgroup of mild AD patients with baseline MMSE  20 (P 5 .0002, while P , .0083 is considered significant after adjusted for multiple comparisons with Bonferroni correction) (Fig. 3B), whereas there is no statistically significant difference in the rate of change of either ADAS-Cog or MMSE scores when grouped by age or gender of the subjects (Figs. 3A, 3C, 3D, and 3F). This suggests that UB-311 may have a potential of cognition improvement in patients with early stage of Alzheimer’s dementia.

web 4C=FPO

728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788

7

Fig. 3. Mean change over time in ADAS-Cog and MMSE scores by age, MMSE at baseline, or gender. For assessment of ADAS-cog and MMSE scores, UB311-treated patients were stratified by (A and D) age in years (,60 and 60), (B and E) MMSE score (,20 and  20), or (C and F) gender (male and female). A positive change from baseline in ADAS-Cog score represents cognitive deterioration. For MMSE, a negative change indicates deterioration. Error bars are standard error. (B) MMSE score (,20 and 20); *P , .0002 for the difference in the rate of disease progression. Abbreviations: ADAS-Cog, Alzheimer’s Disease Assessment Scale–Cognitive Subscale; MMSE, Mini–Mental State Examination. FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849

8

850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

4. Discussion By design, UB-311 vaccine has several safety features. First, site-specific immunogenicity is directed to the N-terminus (Ab1–10) (Table 2) and is devoid of intrinsic Ab-specific T-cell epitopes located between amino acids 16–33 [19], the Ab T-cell sites possibly responsible for the adverse T-cell autoimmunity in the case of AN-1792 (Ab1–42) vaccine [20]. Second, UB-311 is formulated in a Th2-biased delivery system containing CpG ODN, known to favor generalized B-cell mitogenicity with a preference for regulatory Th2 responses rather than Th1 proinflammatory T-cell responses (US patent number 8,088,388); and alum is known to induce Th2-polarized immune responses [21]. The meningoencephalitis associated with AN-1792 vaccine may have also been exacerbated by a Th1-biased adjuvant composition that included QS-21 (saponin) adjuvant and polysorbate-80 surfactant in the formulation [8]. Thus, it is important to design a vaccine product that favors Th2-only while inhibiting without abrogating Th1 immunity [22,23]. Of note, Alzheimer’s disease represents a state of unhealthy aging, termed as “inflamm-aging,” marked by a decline of Th2 responses and chronic upregulation of Th1 proinflammatory mediators such as IL-6, TNF-a, and C-reactive protein in the brain [24,25] and by an inflammation-driven breakdown of blood brain barrier beginning in the hippocampus that could contribute to cognitive impairment [26]. UB-311 is safe and well tolerated in the chronic toxicity study in cynomolgus macaques and in FIH trial with AD patients, where no meningoencephalitis was reported. Of note, neither microglial cell activation nor T-cell infiltration was observed in brain tissue–staining studies with hAPP751 transgenic mice and cynomolgus macaques [18]. Furthermore, lack of PBMC proliferation responses and cytokine secretion in AD patients (Supplementary Tables 4 and 5) indicate that UB-311 did not generate inflammatory antiself, cell-mediated immune responses. The two UBITh T-helper cell peptide epitopes, MvF5 Th and HBsAg3 Th, could maximize both anti-Ab antibody titers and responder rates without generating significant antiepitope antibody responses (Supplementary Table 1). The epitopes are expected to provide broader and stronger Tcell help than the incidental intrinsic T-helper epitopes of aggregated Ab1–42, which may improve immunogenicity in an elderly population. This was observed that UB-311 generated a 100% responder rate for all AD patients in FIH trial (Figs. 2B and 2C; Table 2), and good antibody responses sustained throughout the 48-week study period (Fig. 2A). One additional important feature of the UBITh platform–based vaccine design is that the responses to chimeric UBITh anti-self-immunogens are reversible and can be maintained by booster immunizations [27–30]. Achieving a 100% responder rate is rare in the vaccine development history. AN-1792 vaccine in AD patients (with QS-21 as adjuvant) had a responder rate of ,25% [7,8] but increased to 60% after additional injections with

polysorbate-80 added in the formulation [31]. The CAD106 (Ab1–6) vaccine with a bacteriophage Qb carrier protein had improved anti-Ab antibody response in 82% of AD patients but only for a short period with anti-Qb antibodies still detected [10]. Overall, in contrast with UB311, the current second-generation Ab vaccines were reported with a 20%–80% responder rate, including ACC001, CAD106, V950, affitopes, AD01, and AD02 [9–12]. The goal for active immunotherapy is to induce antibodies specific for both Ab monomers and oligomers. Ab peptides circulate in blood, cerebrospinal fluid, and brain interstitial fluid mainly as soluble Ab1–40 [32,33]. Senile plaques contain both Ab1–40 and Ab1–42 [34], while vascular amyloid is predominantly the shorter Ab1–40. Measurement of plasma Ab1–40 could thus serve as a pharmacodynamic marker. The observation of plasma Ab1–40 in cynomolgus macaques and human subjects (Supplementary Tables 6 and 7) suggests that UB-311 may operate through the mechanism of promoting efflux of Ab peptides from brain, supporting the “amyloid sink hypothesis” [35]. In AD patients, UB-311 can generate antibodies able to bind both Ab1–42 monomers and oligomers (Figs. 2B and 2C); and, in fact, the antibodies preferentially bind to fibrils, followed by oligomers, and weakly to monomers per dotblot analysis (Fig. 2D). This is of considerable clinical interest as amyloid oligomers, rather than plaques, play a more important role for the disease development [36,37]; and Ab1–42 oligomers are most toxic to neurons compared to Ab1–42 monomers or amyloid plaques [1,2,38]. Ab oligomers can contribute to AD pathogenesis by affecting synaptic plasticity and inhibition of long-term potentiation [38]. Of note, Ab aggregates can cause certain inflammatory responses that lead to formation of tau aggregates and neurofibrillary tangles [39]; and oligomeric Ab-induced IL-1b secretion by microglia may also augment neuroinflammation and increase neuronal cell death [40]. The phase-I observations suggest that UB-311 immunization may have a potential of cognition improvement in patients with mild AD. Compared with patients with moderate AD, those with mild AD (MMSE  20) revealed a stable trend in ADAS-Cog scores with a significant slowdown in deterioration and a slower worsening in MMSE (Fig. 3B). These findings indicate that patients with mild AD have a better cognitive response to UB-311 (P 5.0002), and UB-311 may have a positive therapeutic effect in patients with early stage of Alzheimer’s disease. The last two decades have seen robust development of disease-modifying, passive, and active anti-Ab immunotherapy [5,13,14,41,42]; however, there has been very limited convincing clinical benefits of cognitive improvement [13,38]. Bapineuzumab [43] and solanezumab [44] failed to improve cognition or functional ability in patients with mild-to-moderate AD. Bapineuzumab was limited to lower doses, which were believed to be suboptimal for efficacy, due to high incidence of amyloid-related imaging abnormalities–edema [45]. Although potential benefits of both

FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 Q7 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032

antibodies were observed in patients with mild dementia when stratified by disease severity, solanezumab in the recent EXPEDITION3 phase-III trial disappointingly failed to meet the primary end point in mild AD patients (http://www.medscape. com/viewarticle/873143?src5wnl_edit_tpal). The chance to overcome mild AD may still exist. Aducanumab in a phase-Ib trial has shown a tantalizing but tenuous evidence that the mAb dose-dependently reduces Ab plaque that correlates with cognitive improvement [46]. Unlike solanezumab that binds tightly to Ab monomers but not to the more toxic soluble oligomers or insoluble fibrils [47], aducanumab selectively binds aggregated forms of b-amyloid, including soluble oligomers and insoluble fibrils, but does not bind monomers [48]. The cognitive efficacy by aducanumab implies that a similar positive effect may be achieved via active immunotherapy. UB-311 immunotherapy in patients with mild AD patients (Fig. 3B) may be mediated through a target-binding mechanism similar to that by aducanumab, that is, preferential binding to oligomers (Fig. 2C) and fibrils (Fig. 2D). UB-311 binds to Ab monomers as well (Fig. 2B). Overall, the potential of cognition improvement observed with aducanumab [46] also support the amyloid cascade theory [39,48] including Ab-induced tau pathology that underlies the Alzheimer’s disease [49 51]. Based on UB-311’s favorable results from FIH trial (24week treatment plus 24-week observational follow-up), a randomized, double-blind, placebo-controlled, 78-week, multicenter phase-IIa trial (identifier NCT02551809) was initiated in 2015 to characterize longer term safety, tolerability, immunogenicity, and efficacy of two different dose regimens (after the initial 0, 4, and 12 weeks priming doses, additional booster doses are given at 12-week or 24-week interval) in 45 patients with mild AD and a positive Ab PET imaging scan at baseline. The phase-IIa trial is expected to conclude in 2018. No serious drug-related adverse events have been reported to date. Acknowledgments The authors wish to thank the study patients and their family members and the clinical investigators P.-N.W. and M.-J.C. and their site personnel. The FIH studies reported in this study were supported by United Biomedical, Inc., Hauppauge, New York, United Biomedical, Inc., Asia, Taiwan, and the Ministry of Economic Affairs Grant, Taiwan (grant proposal: UBITh AD Immunotherapeutic Vaccine [UB-311] for phase-1 clinical trial; the fund no. 98-EC-17A-20-1-0014). The authors also want to thank Dr. Ruby Chen at Academia Sinica whose laboratory performed the dot-blot analytical assay. Authors’ contributions: C.Y.W. conceived the design of the peptide vaccine and directed the UB-311 vaccine program. P.-N.W. and M.-J.C. were principal investigators and contributed equally to the clinical trial. C.L.F., F.L., X.D.F., K.Z., C.H.H., W.-J.P., J.W., S.L., Y.-H.T., Y.T., and C.-C.Y. designed

9

and performed the experiments and also collected and analyzed output data. C.Y.W., C.L.F., B.S.K., and P.A.F. wrote the article. All authors discussed the results and implications and commented on the article at all stages. All authors contributed extensively to the work presented in this study. C.Y.W., C.L.F., F.L., X.D.F., K.Z., W.-J.P., J.W., S.L., C.H.H., Y.-H.T., Y.T., C.-C.Y., B.-S.K., and P.A.F. are salaried employees and shareholders of the respective companies within the United Biomedical, Inc. group, the developer and owner of the UBITh AD Immunotherapeutic Vaccine (UB-311) and the sponsor of the clinical trials. In addition, C.Y.W. is a cofounder of the respective companies within the United Biomedical, Inc. group. C.Y.W. is an inventor on two filed patents titled “Immunogenic peptide composition as vaccines for the prevention and treatment of Alzheimer’s disease.” US patent nos. 6,713,301/ 7,951,909/8,232,373, United States: United Biomedical Inc., 2005/2011/2012, and “Peptide vaccine for prevention and immunotherapy of dementia of the Alzheimer’s Type.” US patent no. 9,102,752. United States: United Biomedical Inc., 2015. All other authors declare no competing financial interests. Supplementary data Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.trci.2017.03.005.

RESEARCH IN CONTEXT

1. Systematic review: We searched PubMed, ClinicalTrials.gov, and recent literature reviews that track product development for antibodies and vaccines with antiAb disease–modifying features for Alzheimer’s disease (AD). None of these immunotherapeutics, active or passive, in clinical trial has been shown to effectively improve cognition of AD patients; however, that reduced Ab plaque load correlates with improved cognition scores in mild AD patients has been reported in early aducanumab clinical trial. 2. Interpretation: UB-311 represents a new-generation vaccine design that has potential to prevent or delay the onset of Alzheimer disease. The utility of the vaccine, if successful, would be more costeffective and easier for dose administration than an antibody drug for treatment of Alzheimer’s disease. 3. Future directions: UB-311’s preclinical Ab reduction features and favorable early clinical outcome of potential cognition improvement in mild AD patients have led to the ongoing phase-II trial for early-tomild Alzheimer’s dementia.

FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093

10

1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

References [1] Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 2002; 297:353–6. [2] Kayed R, Lasagna-Reeves CA. Molecular mechanisms of amyloid oligomers toxicity. J Alzheimers Dis 2013;33:S67–78. [3] Kumar DK, Choi SH, Washicosky KJ, Eimer WA, Ghofrani J, Leftkowitz A, et al. Amyloid-b peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med 2016;8:340ra72. [4] Soscia SJ, Kirby JE, Washicosky KJ, Tucker SM, Ingelsson M, Hyman B, et al. The Alzheimer’s disease-associated amyloid-betaprotein is an antimicrobial peptide. PLoS One 2010;5:e9505. [5] Qian XX, Hamad B, Dias-Lalcaca G. The Alzheimer disease market. Nat Rev Drug Discov 2015;14:675–6. [6] Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999;400:173–7. [7] Orgogozo JM, Gilman S, Dartigues JF, Laurent B, Puel M, Kirby LC, et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology 2003;61:46–54. [8] Gilman S, Koller M, Black RS, Jenkins L, Griffith SG, Fox NC, et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology 2005;64:1553–62. [9] Pasquier F, Sadowsky C, Holstein A, Leterme GL, Peng Y, Jackson N, et al. Two phase 2 multiple ascending-dose studies of vautide cridificar (ACC-001) and QS-21 adjuvant in mild-to-moderate Alzheimer’s disease. J Alzheimers Dis 2016;51:1131–43. [10] Winblad B, Andreasen N, Minthon L, Floesser A, Imbert G, Dumortier T, et al. Safety, tolerability and antibody response of active Ab immunotherapy with CAD106 in patients with Alzheimer’s disease: randomised, double-blind, placebo-controlled, first-in-human study. Lancet Neurol 2012;11:597–604. [11] Savage MJ, Wu G, McCampbell A, Wessner KR, Citron M, Ling X, et al. A novel multivalent Abeta peptide vaccine with preclinical evidence of a central immune response that generates antisera recognizing a wide range of Abeta peptide species. Alzheimers Dement 2010;6:S142. Abstract No. O3-07-03. [12] Hendrix S, Ellison N, Stanworth S, Tierney L, Mattner F, Schmidt W, et al. Methodological aspects of the phase II study AFF006 evaluating amyloid-beta-targeting vaccine AFFITOPE AD02 in early Alzheimer’s disease—prospective use of novel composite scales. J Prev Alzheimers Dis 2015;2:91–102. [13] Marciani DJ, Ellison N, Stanworth S, Tierney L, Mattner F, Schmidt W, et al. A retrospective analysis of the Alzheimer’s disease vaccine progress—the critical need for new development strategies. J Neurochem 2016;137:687–700. [14] Agadjanyan MG, Petrovsky N, Ghochikyan A. A fresh perspective from immunologists and vaccine researchers: active vaccination strategies to prevent and reverse Alzheimer’s disease. Alzheimers Dement 2015;11:1246–59. [15] Farlow MR, Andreasen N, Riviere ME, Vostiar I, Vitaliti A, Sovago J, et al. Long-term treatment with Ab immunotherapy with CAD106 in mild Alzheimer’s disease. Alzheimers Res Ther 2015;7:23. [16] Vandenberghe R, Riviere ME, Caputo A, Sovago J, Maguire RP, Farlow M, et al. Active Ab immunotherapy CAD106 in Alzheimer’s disease: A phase 2b study. Alzheimers Dement (N Y) 2017;3:10–22. [17] Cummings J, Morstort T, Lee G. Alzheimer’s drug development pipeline: 2016. Alzheimers Dement (N Y) 2016;2:222–32. [18] Wang CY, Finstad CL, Walfield AM, Sia C, Sokoll KK, Chang TY, et al. Site-specific UBIThÒ amyloid-b vaccine for immunotherapy of Alzheimer’s disease. Vaccine 2007;25:3041–52. [19] Monsonego A, Zota V, Karni A, Krieger JI, Bar-Or A, Bitan G, et al. Increased T cell reactivity to amyloid beta protein in older humans and patients with Alzheimer disease. J Clin Invest 2003;112:415–22.

[20] Greenberg SM, Bacskai BJ, Hyman BT. Alzheimer disease’s double edged vaccine. Nat Med 2003;9:389–90. [21] Ghochikyan A, Mkrtichyan M, Petrushina I, Movsesyan N, Karapetyan A, Cribbs DH, et al. Prototype Alzheimer’s disease epitope vaccine induced strong Th2-type anti-Abeta antibody response with Alum to Quil A adjuvant switch. Vaccine 2006;24:2275–82. [22] Pellicano M, Bulati M, Buffa S, Barbagallo M, Di Prima A, Misiano G, et al. Systemic immune responses in Alzheimer’s disease: in vitro mononuclear cell activation and cytokine production. J Alzheimers Dis 2010;21:181–92. [23] Marciani DJ. Alzheimer’s disease vaccine development: a new strategy focusing on immune modulation. J Neuroimmunol 2015; 287:54–63. [24] Giunta B, Fernandez F, Nikolic WV, Obregon D, Rrapo E, Town T, et al. Inflammaging as a prodrome to Alzheimer’s disease. J Neuroinflammation 2008;5:51. [25] Reale M, Iarlori C, Feliciani C, Gambi D. Peripheral chemokine receptors, their ligands, cytokines and Alzheimer’s disease. J Alzheimers Dis 2008;14:147–59. [26] Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015;85:296–302. [27] Wang CY, Chang TY, Walfield AM, Ye J, Shen M, Chen SP, et al. Effective synthetic peptide vaccine for foot-and-mouth disease in swine. Vaccine 2002;20:2603–10. [28] Wang CY, Shen M, Tam G, Fang XD, Ye J, Shen F, et al. Synthetic AIDS vaccine by targeting HIV receptor. Vaccine 2002;21:89–97. [29] Wang CY, Walfield AM, Fang XD, Hammerberg B, Ye J, Li ML, et al. Synthetic IgE peptide vaccine for immunotherapy of allergy. Vaccine 2003;21:1580–90. [30] Wang CY, Walfield AM. Site-specific peptide vaccines for immunotherapy and immunization against chronic diseases, cancer, infectious diseases, and for veterinary applications. Vaccine 2005;23:2049–56. [31] Bayer AJ, Bullock R, Jones RW, Wilkinson D, Paterson KR, Jenkins L, et al. Evaluation of the safety and immunogenicity of synthetic Abeta42 (AN1792) in patients with AD. Neurology 2005;64:94–101. [32] Ghiso J, Frangione B. Amyloidosis and Alzheimer’s disease. Adv Drug Deliv Rev 2002;54:1539–51. [33] Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci 2011; 12:723–38. [34] Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A 1985;82:4245–9. [35] DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM. Peripheral anti-Abeta antibody alters CNS and plasma Abeta clearance and decreases brain A beta burden in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A 2001;98:8850–5. [36] Zhao LN, Long H, Mu Y, Chew LY. The toxicity of amyloid oligomers. Int J Mol Sci 2012;13:7303–27. [37] Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman CW, et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 2003; 300:486–9. [38] Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med 2008; 14:837–42. [39] Stancu IC, Vasconcelos B, Terwel D, Dewachter I. Models of b-amyloid induced Tau-pathology: the long and “folded” road to understand the mechanism. Mol Neurodegener 2014;9:51. [40] Parajuli B, Sonobe Y, Horiuchi H, Takeuchi H, Mizuno T, Suzumara A. Oligomeric amyloid beta induces IL-1b processing via production of ROS: implication in Alzheimer’s disease. Cell Death Dis 2013;4:e975. [41] Wisniewski T, Goni F. Immunotherapeutic approaches for Alzheimer’s disease. Neuron 2015;85:1162–76.

FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215

C.Y. Wang et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions - (2017) 1-11

1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276

[42] Wisniewski T, Drummond E. Developing therapeutic vaccines against Alzheimer’s disease. Expert Rev Vaccines 2016;15:401–15. [43] Salloway S, Sperling R, Fox NC, Blennow K, Klunk W, Raskind M, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med 2014;370:322–33. [44] Siemers ER, Sundell KL, Carlson C, Case M, Sethuraman G, LiuSeifert H, et al. Phase 3 solanezumab trials: secondary outcomes in mild Alzheimer’s disease patients. Alzheimers Dement 2016; 12:110–20. [45] Sperling R, Salloway S, Brooks DJ, Tampieri D, Barakos J, Fox NC, et al. Amyloid-related imaging abnormalities in patients with Alzheimer’s disease treated with bapineuzumab: a retrospective analysis. Lancet Neurol 2012;11:241–9. [46] Sevigny J, Chiao P, Bussiere T, Weinreb PH, Williams L, Maier M, et al. The antibody aducanumab reduces Ab plaques in Alzheimer’s disease. Nature 2016;537:50–6.

11

[47] Bouter Y, Lopez Noguerola JS, Tucholla P, Crespi GA, Parker MW, Wiltfang J, et al. Abeta targets of the biosimilar antibodies of Bapineuzumab, Crenezumab, Solanezumab in comparison to an antibody against N-truncated Abeta in sporadic Alzheimer disease cases and mouse models. Acta Neuropathol 2015;130:713–29. [48] Reiman EM. Attack on amyloid-b protein. Nature 2016;537:36–7. [49] Zempel H, Luedtke J, Kumar Y, Biermat J, Dawson H, Mandelkow E, et al. Amyloid-b oligomers induce synaptic damage via tau-dependent microtubule severing by TTLL6 and spastin. EMBO J 2013; 32:2920–37. [50] Seward ME, Swanson E, Norambuena A, Reimann A, Cochran JN, Li R, et al. Amyloid-b signals through tau to drive ectopic neuronal cell cycle re-entry in Alzheimer’s disease. J Cell Sci 2013; 126:1278–86. [51] Wang Y, Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci 2016;17:5–21.

FLA 5.4.0 DTD  TRCI83_proof  14 April 2017  4:34 pm  ce

1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337