Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Oct 24.
Published in final edited form as: J Med Chem. 2024 Oct 15;67(20):18265–18289. doi: 10.1021/acs.jmedchem.4c01630

Pharmacophore Establishment and Optimization of Saturated 1,6-Naphthyridine-Fused Quinazolinones that Inhibit Meningoencephalitis-Causing Naegleria fowleri

Matthew S Lish 1, Jillian E M McKeon 2, Caroline M Palmentiero 3, Julia M Pomeroy 4, Colm P Roster 5, Ilia A Guzei 6, James C Morris 7, Jennifer E Golden 8
PMCID: PMC11577639  NIHMSID: NIHMS2031916  PMID: 39402986

Abstract

Primary amoebic meningoencephalitis (PAM) is a human brain infection caused by Naegleria fowleri with a 97% mortality rate. Quinazolinones resulting from a Mannich-coupled domino rearrangement were recently identified as inhibitors of the amoeba. Herein, we resolved the effective concentrations for 25 pilot compounds and then, using the Mannich protocol and a key late-stage, N-demethylation/functionalization, we synthesized 53 additional analogs to improve potency, solubility and microsomal stability. We established an antiamoebic quinazolinone pharmacophore, culminating in (±)-trans-57b which featured the best combination of potency, selectivity index, solubility, and microsomal stability. Enantiomeric separation afforded (4aS,13bR)-57b (BDGR-20237) with a 41-fold potency advantage over its enantiomer. ADME and mouse pharmacokinetic profiling for BDGR-20237 revealed high brain penetrance but a limited half-life which did not statistically enhance the mouse survival in a pilot efficacy study. The pharmacophoric model, supported by 88 quinazolinones, several of which exhibit subnanomolar potency, will guide further scaffold optimization.

Graphical Abstract:

graphic file with name nihms-2031916-f0008.jpg

INTRODUCTION

Primary amoebic meningoencephalitis (PAM) is a rare, yet highly lethal, necrotic brain infection that is caused by the free-living amoeba Naegleria fowleri. 1,2 Warm, freshwater ecosystems and soil are hospitable environments in which N. fowleri thrives, sustaining itself on microbiota.3 Potential for human infection occurs when amoeba-contaminated water enters the nasal cavity, typically encountered by children during recreational water activities in lakes or inadequately treated swimming pools and water parks.4 Alternatively, using infected tap water for nasal irrigation3 can introduce the amoeba unwittingly into the nasal mucosa where the amoebic trophozoite form migrates sequentially through the cribriform plate and olfactory bulb before infiltrating the frontal lobes of the brain.4,5 Post-mortem analysis of the brains of PAM patients has shown severe tissue necrosis and hemorrhaging resulting from action of amoeba predation and the host immune response.69

Symptoms of PAM, which include headache, fever, nausea, fatigue, vomiting, confusion, seizures, and coma, mimic what is observed in cases of bacterial or viral meningoencephalitis, thereby often delaying diagnosis and treatment.4 Infection is associated with a > 97% case fatality rate, and death typically results in 3–7 days after symptom onset.8,10 Among the 157 reported cases in the United States between 1962 and 2022, there have been only four survivors,11 and the infection has been documented in at least 39 nations worldwide.4,12 Successful outcomes rely critically on early diagnosis and therapeutic intervention with repurposed drug cocktails (Figure 1).6,1315

Figure 1.

Figure 1.

Drugs used alone or in combination for PAM.

The efficacy of these treatment regimens is largely anecdotal and limited in statistical significance.15 Nonetheless, in the absence of alternatives, amphotericin B is commonly used alone or in combination with fluconazole, rifampicin, or azithromycin.16 Miltefosine has also been used, but due to mixed results, additional studies are needed to determine its efficacy.17 Given the limited success with repurposed drugs, along with the ambiguity surrounding their efficacy and safety, there is a grave need for the development of brain penetrant, nontoxic and efficacious drugs that improve patient survival.

Following the study of a benzylamine series18 that inhibited N. fowleri amoeba, we examined other scaffolds for a parallel development opportunity. Recently, we disclosed a chemical methodology19 that paired a quinazolinone-based Mannich-like reaction with our dual rearrangement protocol termed as a Sequential Quinazolinone-Amidine Rearrangement Strategy (SQuAReS, B → D, Scheme 1).20 An initial library of quinazolinones D bearing a terminal oblique ring fusion was prepared and represented a novel framework. We were delighted to discover that several members of the collection showed >90% inhibition of N. fowleri at 10 μM, thus warranting further studies.19 Here, we shifted from a synthetic methodology driven program to a medicinal chemistry campaign. First, we resolved 50% effective concentrations (EC50 values) against N. fowleri Nf69 for the pilot compounds, and then expanded structure–activity and structure–property relationships (SAR and SPR, respectively) to establish a pharmacophore.

Scheme 1. Mannich-Coupled Sequential Quinazolinone-Amidine Rearrangement Strategy (SQuAReS).

Scheme 1.

Further, we sought to understand diastereomeric and enantiomeric influences on activity while refining potency, physiochemical properties, and other parameters to enable in vivo evaluation. Herein, we describe the synthesis and antiamoebic activity of 88 analogs that exposed pharmacophoric opportunities that were leveraged to afford exceptionally potent inhibitors of N. fowleri. Further, a prototype was profiled for potency against two amoeba strains, ADME, mouse pharmacokinetics, and preliminary in vivo efficacy to establish baseline parameters and guide further optimization.

RESULTS AND DISCUSSION

Chemistry.

Methodology derived compounds predominately featured a N-methyl substituent on the tertiary amine (R2 of D, Scheme 1), so the first analogs were designed with this region preserved. Assembly of the quinazolinone core was done using N-Boc protected amino acids that were prepared from the hydrolysis of lactams 1a–b or protection of amine 3 to afford intermediates 2a–c (Scheme 2). A two step, one-pot dehydrative cyclization, promoted by triphenylphosphite in the presence of N-Boc-amino acids 2a–c, anthranilic acids 4, and neopentylamine delivered quinazolinone salts 5 after N-Boc deprotection. The salts 5 were used in a AgOTf-promoted Mannich-type reaction with N-Cbz aminopropanal to give cyclized products 6. The cis- and trans-isomers were isolated and taken forward as a mixture into the N-Cbz deprotection and dual rearrangement (quinazolinone to amidine formation, followed by a spontaneous intramolecular cyclization, SQuAReS, Scheme 1) to generate (±)-cis-8a-22a, 24a, 26a-31a ring-fused analogs and (±)-trans-8b-22b, 24b, 26b-30b analogs which were separated using normal phase chromatography. Alternatively, brominated intermediate 6 underwent Suzuki coupling to install a phenyl ring of 7 which was subsequently deprotected and basified to initiate the SQuAReS and afford (±)-cis-23a and (±)-trans-23b after chromatographic separation. A late-stage palladium catalyzed cyanation21 of (±)-trans-24b afforded (±)-trans-25b.

Scheme 2. Synthesis of N-Methyl Quinazolinone Analogsa.

Scheme 2.

aReagents and conditions: (a) KOH, EtOH, 100 °C, 24 h, then (b) Boc2O, NEt3, DMAP, 0 °C to rt, 24 h, 70–90%; (c) P(OPh)3, pyr., 130 °C, 1 h; then (d) neopentlyamine, 145 °C 8 h, 20–80%; (e) HCl in dioxane or MeOH, 3 h (75–95%); (f) CbzNH(CH2)2CHO, AgOTf, 4 Å MS, MeCN, μW 150 °C 1 h, 50–90%; (g) PdCl2(dppf), ArB(OH)2, NaOAc, dioxane, H2O, 95 °C, 82% (h) MsOH, HFIP, 50 °C, 3 h, then (i) NEt3, CH3CN, reflux to 150 °C, 4–18 h; 60–90%; (j) Pd2(dba)3, S-Phos, Zn(CN)2, DMF, H2O, μW 120 °C 1 h, 44%.

The most expedient way to integrate nonmethyl substituents on the tertiary amine was to demethylate the parent compounds, followed by NH-functionalization. Hence, (±)-trans-21b and (±)-trans-24b were N-demethylated2224 using 1-chloroethyl chloroformate to afford the corresponding NH products, (±)-trans-32b and (±)-trans-33b (Scheme 3). Notably, only the trans-six-membered, ring-fused isomers were successfully demethylated in this protocol. The cis-six-membered, ring-fused isomers were substantially less reactive under these conditions, showing <25% conversion, while also generating multiple side products. Additional reaction time, temperature and/or equivalents of ACE-Cl did not improve the outcome. The trans-NH compounds were acylated or alkylated to afford (±)-trans-34b-50b, 53b-57b, 59b-62b, 65b. For (±)-trans-51b-52b, 63b-64b, the N-ethanol intermediate was prepared, followed by O-mesylation and amine displacement. Lastly, 58b was prepared through cyanation21 of the chloride, 57b. Selected analogs were separated by chiral HPLC or supercritical fluid chromatography (SFC) to afford individual enantiomers.

Scheme 3. N-Demethylation/Alkylation Synthetic Sequencea.

Scheme 3.

aReagents and conditions: (a) ACE-Cl, DCE, reflux, 14–16 h, then (b) MeOH, reflux, 21 h, 50—80%; (c) RCOCl, NEt3, DCM, 0 to 50 °C, 70–90%; (d) R2X, Et3N, DMF 50–80 °C, 50–80%; (e) MsCl, NEt3, DCE, 0 °C to rt, then (f) NHR3R4, rt to reflux, 17–49%; (g) Pd2(dba)3, S-Phos, Zn(CN)2, DMF, H2O, μW 120 °C 1 h, 50%.

Despite the focused methodological structural scope and lack of dose response data in our past chemistry study, the amoebic percent inhibition data did reveal some early insights. For instance, five-membered, ring-fused analogs and analogs with six-membered morpholino or piperazino ring fusions did not demonstrate appreciable inhibition at 10 μM. However, several analogs of interest had A-ring substituents, so we explored various core phenyl ring substitutions first (Table 1). Briefly, compounds were assessed in triplicate at multiple concentrations up to 10 μM against N. fowleri Nf69 to generate EC50 values. Miltefosine, a positive control, inhibited the amoeba with an EC50 = 35.7 ± 9.10 μM, a value similar to previously reported data.25 Compounds were also tested at 8 concentrations up to 20 μM using human neuronal SH-SY5Y cells to evaluate toxicity.

Table 1.

A-Ring Substitution and Effect of Cis- or Trans-4a,13b Ring Fusion on Anti-Amoeba Activity

graphic file with name nihms-2031916-t0009.jpg
entry cmpd ring fusion R1 R2 R3 R4 EC50 μMa CC50 μMb
1 ±−8a cis H H H H >10 >20
2 ±−8b trans >10 >20
3 ±−9a cis CH3 H H H >10 >20
4 ±−9b trans >10 >20
5 ±−10a cis Cl H H H >10 >20
6 ±−10b trans >10 >20
7 ±−11a cis H CH3 H H 2.8 ± 0.3 >20
8 ±−11b trans 3.1 ± 0.5 >20
9 ±−12a cis H OCH3 H H >10 >20
10 ±−12b trans 7.3 ± 1.2 >20
11 ±−13a cis H CN H H 2.1 ± 0.3 >20
12 ±−13b trans 0.67 ± 0.05 >20
13 ±−14a cis H F H H >10 >20
14 ±−14b trans >10 >20
15 ±−15a cis H CF3 H H 0.95 ± 0.08 >20
16 ±−15b trans 1.1 ± 0.3 >20
17 ±−16a cis H Cl H H 2.2 ± 0.2 >20
18 ±−16b trans 1.0 ± 0.3 >20
19 ±−17a cis H H OCH3 H 1.8 ± 0.3 >20
20 ±−17b trans 1.5 ± 0.7 >20
21 ±−18a cis H H F H >10 >20
22 ±−18b trans >10 >20
23 ±−19a cis H H CF3 H 6.9 ± 1.8 >20
24 ±−19b trans 0.99 ± 0.08 >20
25 ±−20a cis H H Cl H >10 >20
26 ±−20b trans 6.0 ± 1.7 >20
27 ±−21a cis H Cl Cl H 0.26 ± 0.01 >20
28 ±−21b trans 0.20 ± 0.01 >20
29 ±−22a cis H CF3 Cl H 0.27 ± 0.12 >20
30 ±−22b trans 0.34 ± 0.07 >20
31 ±−23a cis H Cl Ph H 3.4 ± 0.6 >20
32 ±−23b trans 2.1 ± 0.6 9.4
33 ±−24a cis H Cl OCH3 H 0.38 ± 0.03 >20
34 ±−24b trans 0.43 ± 0.14 >20
35 ±−25b trans H CN OCH3 H 3.8 ± 1.1 >20
36 ±−26a cis H Cl H OCH3 >10 >20
37 ±−26b trans >10 >20
38 miltefosine 35.7 ± 9.10 NT
a

Averaged data from triplicate runs (n = 3) with ± SEM using N. fowleri (Nf69 strain, ATCC 30215) trophozoites.

b

Human neuronal SH-SY5Y cells (ATCC CRL-2266); NT = not tested.

The absence of any core substituent other than hydrogen atoms resulted in poor inhibition (Table 1, entry 1, EC50 > 10 μM), regardless of a cis- or trans-ring fusion. Methyl group or chlorine atom substitution at R1 gave the same results. Substitution at R2 alone (entries 7–11) revealed improved, single digit micromolar activity in several cases. A fluorine atom rendered the compound inactive (entries 13–14), and a methoxy group in this position afforded marginal activity for the trans-isomer while the cis isomer was inactive (entries 9–10). A chlorine atom or trifluoromethyl substituent reached similar potency for either diastereomer, in the range of EC50 ~ 1–2 μM (entries 15–18). A nitrile group in this position showed a 3-fold preference over its trans-isomer (entries 11–12). Notable outcomes for R3 included a 7-fold preference for a trifluoromethyl substituent on the trans-ring fused scaffold versus its corresponding cis-isomer and a loss of activity for fluorination at this position.

Chlorine substitution at R2 and R3 afforded a significant potency improvement (EC50 ~ 200 nM range, entries 27–28, Table 1). Similar results were also observed for chlorine combined with trifluoromethyl or methoxy groups. Generally, substitution of the R2 and/or R3 positions with electron withdrawing groups other than fluorine gave better potency with some trending preference for the trans-isomers; however, for the most potent compounds, the diastereomeric preference was negligible. Few examples featuring R4 substituents were prepared due to limited success of the Mannich reaction when R4 was a chlorine or bromine atom. Nonetheless, methoxy substituent at R4 led to poor amoeba inhibition, and in combination with an R2 chloride atom, the pairing was unfavorable (entries 36–37). Except for phenyl substituted derivative, (±)-trans-23b, none of the compounds showed cytotoxicity in neuronal cells up to the highest tested concentrations of 20 μM.

Other skeletal modifications were surveyed (Figure 2). Pyridyl derivatives (±)-cis-27a and (±)-trans 27b were inactive (EC50 > 10 μM). Analogs integrating the favored chlorinated or trifluoromethylated core with other changes included the ring-fused morpholino analogs (±)-cis-28a30a and (±)-trans-28b30b. The 5-membered, ring-fused analog 31a, isolated exclusively as a cis-isomer, was also prepared; however, none of the structural changes in this analog set showed antiamoebic activity or mammalian cytotoxicity when tested up to the highest concentration in each assay (EC50 > 10 μM, CC50 > 20 μM).

Figure 2.

Figure 2.

Summary of analogs with heteroatom insertions or ring contractions made to the quinazolinone framework.

Early in the SAR development, we zeroed in on the dichloro-substitution of quinazolinones 21ab due to the potency gained. In this case, the cis and trans isomers demonstrated virtually the same potency, but for other 6-membered, ring-fused compound pairs, there was an emerging trend that the trans isomer was preferred. Synthetically, the trans-isomer was generally produced in a greater yield than the corresponding cis-product, leading to a deficit of cis-isomer to work with, and when six-membered ring-fused analogs were subjected to the N-demethylation protocol (Scheme 3, conditions a,b), only the trans-isomers reacted productively to afford NH intermediates that could be further derivatized. Taken together, we used exclusively the trans-N-methyl products as starting materials to investigate the effects of other tertiary amine substituents. Dichloroquinazolinone (±)-trans-21b was selected as the initial framework for this survey (Table 2).

Table 2.

Effect of Changing the 3° Amine Substituent on (±)-Trans-Dichloroquinazolinones

graphic file with name nihms-2031916-t0010.jpg

entry cmpd R1 EC50 (μM)a cc50 μMb (SI)c MLM T1/2, mind entry cmpd R1 EC50(μM)a CC50 μMb (SI)c MLM T1/2, mind

1 ±-trans-21b CH3 0.20 ± 0.01 >20 ND 13 ±-trans-44b graphic file with name nihms-2031916-t0011.jpg >10 >20 ND
2 ±-trans-32b H 1.60 ± 0.18 >20 ND 14 ±-trans-45b graphic file with name nihms-2031916-t0012.jpg >10 >20 ND
3 ±-trans-34b graphic file with name nihms-2031916-t0013.jpg 0.040 ± 0.003 >20 41.1 15 ±-trans-46b graphic file with name nihms-2031916-t0014.jpg 0.72 ± 0.08 >20 ND
4 ±-trans-35b graphic file with name nihms-2031916-t0015.jpg 0.007 ± 0.002 >20 19.7 16 ±-trans-47b graphic file with name nihms-2031916-t0016.jpg 0.30 ± 0.02 >20 ND
5 ±-trans-36b graphic file with name nihms-2031916-t0017.jpg 0.002 ± 0.001 4.8 (2400) 18.3 17 ±-trans-48b graphic file with name nihms-2031916-t0018.jpg 0.020 ± 0.02 >20 83.1
6 ±-trans-37b graphic file with name nihms-2031916-t0019.jpg 0.72 ± 0.05 >20 ND 18 ±-trans-49b graphic file with name nihms-2031916-t0020.jpg 0.023 ± 0.009 10.0 (435) 17.0
7 ±-trans-38b graphic file with name nihms-2031916-t0021.jpg 2.53 ± 0.58 >20 ND 19 ±-trans-50b graphic file with name nihms-2031916-t0022.jpg 0.0004 ± 0.0001 >20 9.8
8 ±-trans-39b graphic file with name nihms-2031916-t0023.jpg 0.0002 ± 0.0001 >20 21.9 20 ±-trans-51b graphic file with name nihms-2031916-t0024.jpg 0.18 ± 0.04 8.0 (44) ND
9 ±-trans-40b graphic file with name nihms-2031916-t0025.jpg 0.48 ± 0.13 >20 ND 21 ±-trans-52b graphic file with name nihms-2031916-t0026.jpg 0.13 ± 0.02 6.4 (49) ND
10 ±-trans-41b graphic file with name nihms-2031916-t0027.jpg 0.05 ± 0.01 >20 ND 22 ±-trans-53b graphic file with name nihms-2031916-t0028.jpg 0.006 ± 0.001 >20 109.0
11 ±-trans-42b graphic file with name nihms-2031916-t0029.jpg >10 >20 ND 23 ±-trans-54b graphic file with name nihms-2031916-t0030.jpg 0.007 ± 0.002 >20 67.2
12 ±-trans-43b graphic file with name nihms-2031916-t0031.jpg >10 >20 ND 24 ±-trans-55b graphic file with name nihms-2031916-t0032.jpg 0.011 ± 0.001 >20 75.2
a

Averaged data from triplicate runs (n = 3) with ± SEM using N. fowleri (NF69 strain, ATCC 30215) trophozoites; miltefosine (positive control) N. fowleri EC50 = 35.7 ± 9.1 0 μM.

b

Human neuronal SH-SY5Y cells (ATCC CRL-2266).

c

SI = selectivity index, CC50/EC50.

d

Metabolic stability in CD-1 strain mouse liver microsomes. ND – not determined.

The NH intermediate (±)-trans-32b showed an 8-fold loss in antiamoebic potency compared to the parent N-methyl analog, (±)-trans-21b, from which it was derived. Fortunately, several other appendages attached to this nitrogen atom resulted in spectacular potency (Table 2). For instance, installing an N-isobutyl group or a methylene-bridged cyclopropyl group registered EC50 values of 7 and 2 nM, respectively (entries 4–5, Table 2), though some cytotoxicity was noted for the latter compound (selectivity index (SI) = 2400). A benzyl group was also well tolerated, but a scan of methylene-linked pyridyl groups revealed a trend in preference of 3 pyridyl ≫ 4-pyridyl ≫ 2-pyridyl in which the 3-pyridyl analog demonstrated an astonishing EC50 of 0.2 nM (entry 8). The N-acylated derivatives bearing a bulky α substituent to the carbonyl group lost activity (11–14); however, submicromolar activity was restored with the less bulky dimethylaminoalkylamide (±)-trans-46b. The alkyl tethered alcohol and ethers (entries 17–19 and 22–24) were noteworthy performers with several landing in the single digit nanomolar potency range. Select analogs from this group were assessed for microsomal stability in mice before proceeding to additional structural refinements. Several of the most potent compounds showed suboptimal microsomal stability with half-lives of <60 min (entries 3–5, 8, 18–19). While (±)-trans-48b passed this benchmark with a half-life of 83 min, its kinetic solubility was limited (<10 μM). The morpholino derivative, (±)-trans-55b, was more soluble (206 μM), but still exhibited marginal microsomal stability (entry 24). Oxetane (±)-trans-53b was potent and showed the best stability of the tested analogs with a half-life of 109 min, though solubility was determined to be 40 μM (entry 22). Nonetheless, these were attractive structural starting points from which enhanced solubility and microsomal stability was expected to emerge with further optimization. As such, we sought to exchange one of the two aryl chlorine atoms with substituents that were anticipated to improve the profile.

We used oxetane (±)-trans-53b as a starting point for refinement (entry 10, Table 3). Exchange of the C11 chlorine atom with a methoxy group afforded (±)-trans-57b with an EC50 of 29 nM, solubility of 198 μM, and microsomal stability half-life of >145 min. A combination of nitrile and methoxy groups (entry 14) was investigated due to the noted benefit on microsomal stability and lipophilic efficiency when replacing an aromatic chloride with an aromatic nitrile.26 These changes, when paired with the oxetane, showed promising attributes, though they did not outperform the chloro/methoxy modification. Some of the best performing tertiary amine substituents assessed in earlier analogs, along with several others that reduced aliphatic carbon count or introduced solubilizing groups, were also surveyed. However, the combination of potency, solubility, and microsomal stability favored (±)-trans-57b.

Table 3.

Effects of A-Ring Substitution and 3° Amine Substituent on Potency, Solubility and Microsomal Stability and Resolution of Selected Racemic Analogs to Compare Enantiomeric Data

graphic file with name nihms-2031916-t0033.jpg

entry cmpd R1 R2 R3 EC50(μM)a CC50 μMb SIc solubility μM MLM, T1/2, mind

1 ±-trans-15b 1.1 ± 0.3 >20 >18.2 159 21.2
2 trans-15b-ent-1 CF3 H CH3 0.25 ± 0.02 >20 >80.0 109 10.9
3 trans-15b-ent-2 4.3 ± 2.3 >20 >4.7 ND ND

4 ±-trans-16b 1.0 ± 0.3 >20 >20.0 187 18.5
5 trans-16b-ent-1 Cl H CH3 0.28 ± 0.01 >20 >71.4 129 11.5
6 trans-16b-ent-2 4.0 ± 2.6 >20 >5.0 ND ND

7 ±-trans-21b 0.20 ± 0.01 >20 >100 138 ND
8 trans-21b-ent-1 Cl Cl CH3 0.086 ± 0.005 7.8 90.7 98.2 6.1
9 trans-21b-ent-2 2.6 ± 0.2 >20 >7.7 ND ND

10 ±-trans-53b Cl Cl graphic file with name nihms-2031916-t0034.jpg 0.006 ± 0.001 >20 >33000 40.0 109.0
11 ±-trans-57b 0.029 ± 0.003 5.0 172 198 >145
12 trans-57b-ent-1 BDGR-20236 Cl OCH3 graphic file with name nihms-2031916-t0035.jpg 0.49 ± 0.081 >20 >40.8 29.1 75.6
13 trans-57b-ent-2 BDGR-20237 0.012 ± 0.001 2.3 191.7 23.9 48.2

14 ±-trans-58b CN OCH3 graphic file with name nihms-2031916-t0036.jpg 0.12 ± 0.02 >20 >166.7 136 104.6

15 ±-trans-59b Cl OCH3 graphic file with name nihms-2031916-t0037.jpg 0.015 ± 0.001 0.64 42.7 4.6 3.8

16 ±-trans-60b Cl OCH3 graphic file with name nihms-2031916-t0038.jpg 0.023 ± 0.003 1.0 43.5 76.1 12.6

17 ±-trans-61b Cl OCH3 graphic file with name nihms-2031916-t0039.jpg 0.11 ± 0.01 2.5 22.7 65.7 3.1

18 ±-trans-62b 0.17 ± 0.04 1.8 10.6 ND ND
19 trans-62b-ent-1 Cl OCH3 graphic file with name nihms-2031916-t0040.jpg 0.62 ± 0.30 >20 >32.3 ND ND
20 trans-62b-ent-2 0.027 ± 0.003 1.0 38.0 2.6 ND

21 ±-trans-63b Cl OCH3 graphic file with name nihms-2031916-t0041.jpg 0.52 ± 0.06 >20 >38.5 43.1 29.8

22 ±-trans-64b Cl OCH3 graphic file with name nihms-2031916-t0042.jpg 0.74 ± 0.25 >20 >27.0 172 >145

23 ±-trans-65b Cl OCH3 graphic file with name nihms-2031916-t0043.jpg 0.12 ± 0.01 >20 >166.7 3.8 ND

24 miltefosine 35.7 ± 9.10 ND ND ND ND
a

Averaged data from triplicate runs (n = 3) with ± SEM using N. fowleri (Nf69 strain, ATCC 30215) trophozoites.

b

Human neuronal SH-SY5Y cells (ATCC CRL-2266).

c

SI = selectivity index, CC50/EC50.

d

Metabolic stability in CD-1 strain mouse liver microsomes. ND = not determined.

We had mainly been working with racemic analogs, as chiral separation proved challenging; however, we were able to spot check the behavior of selected, enantioenriched (>96–99%) compounds that were separated (Table 3). Chiral prep HPLC conditions were identified for several early analogs featuring an N-methyl substituent, including (±)-trans-15b, (±)-trans-16b, and (±)-trans-21b. In all cases in which the enantiomers were resolved for a given compound, the isomer with the shortest retention time was given a suffix of “ent-1” while the isomer with the longer retention time was labeled ‘ent-2’. These three compounds, separated on the same chiral column matrix, showed that the enantiomer with the shortest retention time was substantially more active than the later eluting isomer (entries 2/3, 5/6 and 8/9, Table 3). For (±)-trans-21b, a >30-fold difference was noted for the more active enantiomer. Evaluation of the most potent enantiomers for these three early analogs showed impaired MLM stability which was a focus of further optimization. For (±)-trans-62b, a different chiral column was employed successfully, revealing that the late-eluting enantiomer was the more potent of the two; however, suboptimal microsomal stability was observed for this analog, too. Despite several attempts to use standard chiral HPLC to separate the enantiomers of one of the best performing analogs, (±)-trans-57b, complete resolution could not be achieved with our techniques and equipment. Ultimately, the separation was successfully performed using chiral SFC.

With separated isomers in hand, trans-57-ent-2 was shown to have a 41-fold advantage in potency over the other isomer (EC50 = 12 nM, entry 13, Table 3). Solubility and microsomal stability were reduced compared to the racemate; however, the values were reasonable to work with in terms of benchmarking spectrum of activity across another N. fowleri strain and establishing physiochemical and initial ADME parameters. In parallel, each enantiomer was individually crystallized and analyzed by single-crystal X-ray crystallography to unequivocally establish the respective absolute configuration (Figure 3). The less active isomer, trans-57b-ent-1, designated BDGR-20236 internally, was determined to possess the 4aR,13bS-configuration at the ring fusion while the more potent isomer, trans-57b-ent-2 (BDGR-20237) had the 4aS, 13bR-configuration.

Figure 3.

Figure 3.

X-ray derived molecular drawings of resolved enantiomers (4aR,13bS)-57b-ent-1 (BDGR-20236) and (4aS,13bR)-57b-ent-2 (BDGR-20237), each bottom panel shown with 50% probability ellipsoids.

Both enantiomers of trans-57b were evaluated against N. fowleri TY strain, isolated from a patient in Virginia, to assess potential differences in susceptibility compared to Nf69 (origin: Australia) which may be genetically distinct (Table 4). Exquisite congruency was observed for both enantiomers across the two strains. Physiochemical and ADME parameters were then benchmarked. The obtained log D value of 2.3 was within an ideal range for drug-likeness. Kinetic aqueous solubility was modest at 24–29 μM.

Table 4.

Strain Potency, Physiochemical, and ADME Data

BDGR-20236 (4aR,13bS)-57b-ent-1 BDGR-20237 (4aS,13bR)-57b-ent-2
N. fowleri EC50, μM Nf69 straina 0.49 ± 0.081 0.012 ± 0.001
TY strainb 0.47 ± 0.096 0.014 ± 0.002
log D7.4c 2.3 2.3
solubilityd 29 μM 24 μM
BBB PAMPA, Peavg (nm/s)e 182, CNS+ 172, CNS+
MLM, T1/2f 75.6 min 48.2 min
HLM, T1/2 80.3 min 92.7 min
plasma stability, T1/2f,g >289 min >289 min
plasma protein bindingf264 74% bound 74% bound
a

N. fowleri (Nf69 strain, ATCC 30215) trophozoites.

b

N. fowleri (TY strain, ATCC 30107) trophozoites; Averaged data from triplicate runs (n = 3) with ± SEM.

c

In octanol/phosphate buffer, pH 7.4.

d

Kinetic solubility, 50 mM phosphate buffer.

e

Averaged data from duplicate runs, 4 h; using positive and negative controls, Pe > 15 nm/s = CNS+; Pe ≤ 15 = CNS-.

f

CD-1 mouse.

g

Microsomal stability expressed in half-life, 60 min exposure, +NADPH.

We assessed the likelihood of brain penetration using an in vitro blood–brain barrier (BBB) parallel artificial membrane permeability assay (PAMPA),27 results of which predicted excellent BBB permeability compared to controls. The MLM stability was better for the less active isomer, BDGR-20236; however marginally, by 27 min. Mouse plasma stability was superb, and a significant free fraction (26%) was observed in mice. Taken together, BDGR-20237, while not ideal in terms of solubility and metabolic stability, was deemed suitable for a mouse pharmacokinetic (PK) study to assess exposure in plasma and brain tissue. Mice (n = 3/group) were dosed independently with a single bolus of BDGR-20237 (11 mg/kg) by either intravenous (IV) or oral (PO) delivery. Plasma concentrations were assessed at six time points over 12 h, and brain concentrations were determined at the experimental end point. Collected PK parameters (Table 5) revealed a half-life of ~1 h by either route and a linear decrease in plasma exposure over 12 h in both the IV and PO dosed cohorts with bioavailability determined to be 38%.

Table 5.

PK Parameters for BDGR-20237 in CD-1 Micea

parameter dose mg/kgb route resultc
half-life, T1/2 (h)d 11 IV 1.0 ± 0.2
PO 1.1 ± 0.4
Vd (L/kg)d 11 IV 3.7 ± 0.2
Cl (mL/min/kg)d 11 IV 40.8 ± 7.6
plasma AUCinf (ng/h/mL)d 11 IV 5011 ± 857
PO 1920 ± 194
plasma conc. at single time points (ng/mL) 0.5 hd 11 IV 2113 ± 238
PO 469 ± 81
2 hd 11 IV 882 ± 189
PO 459 ± 109
4 hd 11 IV 281 ± 110
PO 254 ± 18
8 hd 11 IV 18.1 ± 15.0
PO 21.1 ± 5.5
12 hd 11 IV 2.6 ± ND
PO 2.0 ± ND
2h 44 IP 3849 ± 390
4h 44 IP 1917 ± 409
brain conc. (ng/g)e 12 hd 11 IV or PO low to BLDg
4h 44 IP 9089 ± 2392
B/P ratiof 4h 44 IP 4.7
bioavailability, %Fd 11 PO 38%
a

Female CD-1 mice, n = 3/group.

b

Actual dose reported rather than intended 10 or 50 mg/kg doses.

c

Averaged data from triplicate runs with standard deviation reported.

d

Data from 12 h experiment.

e

Brain homogenate.

f

Brain-to-plasma ratio obtained from the ratio of brain concentration divided by plasma concentration, each respective data point at 4 h.

g

BLD = below levels of detection for two of three mice in each group; brain concentrations for: one mouse in IV group: 13.3 ng/mL; one mouse in PO group: 10.4 ng/mL.

Using a 11 mg/kg dose, the concentration of BDGR-20237 in brain tissue at 12 h was shown to be near or below the limits of detection (BLD). To verify brain penetration, a follow up PK experiment in mice (n = 3/group) was done with a single intraperitoneal (IP) bolus of BDGR-20237 at a dose of 44 mg/kg with plasma concentrations determined at 2 and 4 h. Brain exposure was determined at 4 h (Table 5). We observed that increasing the dose by 5-fold (IP dosing) compared to the first experiment boosted the plasma exposure such that at the 4 h time point, exposure was ~8-fold higher than that observed with IV or PO dosing at the lower dose. Furthermore, at 4 h we observed high brain exposure with a brain-to-plasma ratio (B/P) of 4.7.

Considering these outcomes, the efficacy of BDGR-20237 was assessed using female CD-1 mice which were intranasally infected with N. fowleri Nf69 trophozoites. The study design included three groups of mice (n = 6/group) that were infected on day 0 (Figure 4, panel A). After a 48 h induction period to permit trophozoites to establish brain infection (days 0–2), separate cohorts of mice were treated for 5 days via IP administration using amphotericin B (5 mg/kg, once a day, QD) or BDGR-20237 (11 mg/kg, twice daily, BID). A control group of infected mice received only the vehicle during the treatment window. The 14-day study concluded with the brain tissue of surviving mice being cultured for viable amoeba.

Figure 4.

Figure 4.

(A) Study design of mouse efficacy study using N. fowleri Nf69. (B) Kaplan–Meier survival data for infected mice treated with amphotericin (B, C) Kaplan–Meier survival data for infected mice treated with BDGR-20237.

The onset of neurological symptoms in the infected mice occurred more rapidly than typically observed, as the pathogenicity of the cultured amoeba can be challenging to consistently calibrate at the inception of the study. The amoeba was lethal, resulting in the expiration of all infected mice lacking treatment by day 6 of the experiment. Two of the six amphotericin B treated mice survived and exhibited normal behavior. Upon assessment of brain tissue from the two survivors, amoeba was detected in one. Resultantly, survivability of the amphoB treated group was 17% (Figure 4, panel B). Infected mice treated with BDGR-20237 succumbed to infection by day 7 (Figure 4, panel C). Neither of the two treatment groups (i.e., amphoB or BDGR treated animals), despite the survivors from the amphotericin treated cohort, provided statistically significant extension of life over vehicle controls, though both treatment groups were “trending” toward significance (p value >0.1). Complications from the rapid disease onset may have obscured this value. Nonetheless, considering the ADME and PK profiles for BDGR-20237, along with the pilot mouse efficacy assessment, it is likely that the 11 mg/kg BID dosing over 5 days was insufficient to maintain adequate brain concentrations to provide protection to infected mice. While the 50 mg/kg PK study comparatively revealed much greater plasma and brain exposure and mice did not show any clinical signs of adverse events after administration of a single bolus, the brain concentration of the compound was high at this dose (est. ~20 μM), and there was concern that repeat dosing at this concentration may lead to toxicity given the cytotoxicity that was observed with neuronal cells. Consequently, an assessment of the maximum tolerated dose (MTD) will be performed to guide optimal dosing in future efficacy studies.

CONCLUSIONS

Although rare, the human brain infection caused by N. fowleri is highly lethal. Repurposed drugs, though limited in successful intervention, are frequently used to combat the infection due to lack of investment in the discovery and development of safe and effective CNS-penetrant drugs specifically designed to address PAM. Out of a recent synthetic methodology aimed at constructing novel, pharmacologically biased heterocyclic frameworks, we fortuitously discovered inhibitory activity against the N. fowleri amoeba. These quinazolinones contain an oblique ring fusion with an optimally positioned nitrogen atom, signifying a new asset in the bleak arsenal of scaffolds that show promise against the pathogen. Implementation of the Mannich coupled domino rearrangement and execution of a key late stage N-demethylation/functionalization procedure enabled efficient analog diversification, resulting in the design and synthesis of an 88-member compound collection. These quinazolinones defined a pharmacophoric model from which to optimize several tunable regions. Exceptionally potent analogs with subnanomolar activity were identified, and as is most often the case, compound advancement depended critically on a comprehensive balance of potency, cytotoxicity, solubility, and microsomal stability. From this effort, a single enantiomer, BDGR-20237, exhibited equivalent double-digit nanomolar potency for two wild type strains of N. fowleri, and though some cytotoxicity was noted in a human neuronal cell line, the selectivity index of 192 provided a reasonable window to benchmark mouse PK parameters at two doses and confirm brain exposure. Despite some limitations in ADME and PK, a pilot study of BDGR-20237 was performed in a mouse infection model, revealing that further refinements are necessary, and a strategy for dosing frequency and duration will be required. As these compounds emerged from a phenotypic screen, studies are underway to elucidate a mechanism of action for this chemical series. Nonetheless, this new quinazolinone prototype represents a promising development scaffold that will enhance our understanding of N. fowleri vulnerability, thereby generating opportunity for advancement of new therapeutics.

EXPERIMENTAL SECTION

General Chemistry Experimental Section.

The purity of all final compounds was confirmed to be ≥95% by HPLC/MS. Commercial reagents and solvents were purchased from Alfa Aesar, Ambeed, Combi-Blocks, Fischer Scientific, VWR International, Chem-Impex Int’l, Inc., Acros Organics, etc. Chromatographic purifications were accomplished using a Teledyne Isco CombiFlash Rf 300 purification system with either Redi-sep silica gel columns (normal-phase) or Redi-sep gold C18 columns (reverse phase). LCMS/HPLC analysis was used to monitor reactions and determine purity and was performed on an either an Agilent 1290 Infinity II HPLC system with a 1290 Infinity II Diode Array Detector and an Agilent 6120 Quadrupole LC-MS system with the following parameters: Poroshell 120 EC-C18, 1.9 μm column, UV detection wavelength = 254 nm, flow rate = 1.0 mL/min, gradient = 20–100% LC-MS grade methanol over 4 min; the organic mobile phase and aqueous mobile phase contained 0.1% LC-MS grade formic acid or an Shimadzu LCMS2020 system with a SPD-M40 photodiode array detector with the following parameters: Shim-Pack Velox SP-C18 2.7 μm column, UV detection wavelength = 254 nm, flow rate = 1.0 mL/min, gradient = 20–100% LC-MS grade methanol over 4 min; the organic mobile phase and aqueous mobile phase contained 0.1% LC-MS grade formic acid. Preparative HPLC chiral separations were performed on an ACCQPrep HP150 using either a Regis Reflect I-Cellulose B (5 μm (3,5-diphenylcarbamate), 25 cm × 21.1 mm) column or a Regis (R,R)-Welk-O 1 (5 μm Kromasil, 25 cm × 21.1 mm) column. Analysis of enantiopurity was done using a chiral phase HPLC (Agilent 1200- Series HPLC) wither either a Reflect ICellulose B5 μm (3,5-diphenylcarbamate), 25 cm × 21.1 mm column or a (R,R)-Welk-O 1 5 μm Kromasil, 25 cm × 21.1 mm column. Microwave irradiated (MWI) reactions were carried out using a Biotage Initiator+ Fourth Generation microwave synthesizer. High resolution mass spectra (HRMS) were obtained by the Analytical Instrument Center at the School of Pharmacy on an Electron Spray Injection (ESI) mass spectrometer. 1H, 19F, and 13C nuclear magnetic resonance (NMR) spectra were obtained on either a Varian 500 MHz or a Bruker Ascend 400 MHz spectrometer in chloroform-d. DMSO-d6, D2O, or acetic-acid-d6. The chemical shifts (δ) reported are given in parts per million (ppm). The following abbreviations were used to describe peak splitting patterns: s = singlet, bs = broad singlet, d = doublet, t = triplet, q = quartet, p = pentuplet, dd = doublet of doublet, dt = doublet of triplet, dq = doublet of quartet, td = triplet of doublet, tt = triplet of triplet, qd = quartet of doublet, ddd = doublet of doublet of doublet, tdd = triplet of doublet of doublet, dddd = doublet of doublet of doublet of doublet and m = multiplet. Coupling constants, J, which were reported in Hertz (Hz). The following compounds were previously reported, and the analytical data obtained matched what was previously reported:19 intermediates 2a–c, and final compounds: (±)-cis-8–9a, (±)-cis-11–14a, (±)-cis-16–20a, (±)-cis-27–28a, (±)-trans-8–9b, (±)-trans-11–14b, (±)-trans-16–20b, (±)-trans-27–28b.

General Procedure 1: Synthesis of N-Boc-Protected Quinazolinones.

Substituted 2-aminobenzoic acid 4 (1 equiv), N-Boc-protected amino-acid 2 (1.2 equiv), and P(OPh)3 (2.2 equiv) were combined in anhydrous pyridine (0.12 M). After heating thermally at 130 °C in a pressure relief vial for 1 h, the reaction was cooled to rt, and neopentyl amine (2 equiv) was added. The reaction mixture was heated thermally to 145 °C for 8 h. After cooling, solvent was removed under reduced pressure. The crude oil was dissolved in EtOAc (80 mL), and sequentially washed with 10% citric acid, sat. aq NaHCO3, and brine. The separated organic extract was dried over Na2SO4, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash chromatography to afford S1–S10.

General Procedure 2: Synthesis of Hydrochloride Salts 5a–5j from N-Boc-Protected Quinazolinones.

In a round-bottom flask, the N-Boc-protected quinazolinone S1–S10 (1 equiv) was suspended in either 4 N HCl in dioxane (0.16 M) or 3 N HCl in MeOH (0.16 M) for 2–3 h or until TLC or LCMS showed starting material consumption. The reaction mixture was poured into Et2O and stirred for 10 min. The product HCl salt was then collected by vacuum filtration and dried in a vacuum oven at 60 °C to afford 5a–5j.

General Procedure 3: Synthesis of Mannich-Cyclized Products 6a–6j.

HCl salts 5 (1 equiv), benzyl (3-oxopropyl)carbamate (4 equiv), AgOTf (1 equiv) and activated/Ar-cooled 4 Å MS were placed in an oven-dried 2–5 or 10–20 mL μW vial. The vessel was sealed, evacuated, and backfilled twice with Ar (g), followed by the addition of degassed anhydrous CH3CN (0.15 M). The reaction mixture was then heated by μW irradiation at 150 °C for 1 h. The reaction was filtered over Celite, and the collected organic filtrate was concentrated, then diluted with 20% IPA/CHCl3 (65 mL) and washed with sat. aq NaHCO3 (50 mL). The separated organic extract was dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was suspended in MeOH (10 mL) and absorbed to a 10 g HF-Mega BE-SCX column (Agilent). After rinsing with 3 × 60 mL of MeOH, the absorbed crude material was obtained by washing with 7N NH3 in MeOH (60 mL). After concentration, the residue was purified by flash chromatography to afford 6a–6j.

General Procedure 4: Suzuki Cross-Coupling of Mannich-Cyclized Intermediate to Afford 7.

Mannich product 6e was placed in a 20–40 mL oven-dried pressure relief vial with Pd(dppf)2Cl2 (0.1 equiv), NaOAc (4.5 equiv) and PhB(OH)2 (1.5 equiv). The vial was sealed, evacuated, and backfilled with Ar (g). Degassed anhydrous dioxane/water (3:1) was added, and the vial was heated to 95 °C for 24 h. The reaction mixture was diluted with DCM, washed with H2O, followed by brine. The separated organic extract was dried over Na2SO4, filtered, and concentrated, affording a residue that was purified by normal phase chromatography to afford Suzuki product 7.

General Procedure 5: N-Cbz Deprotection and Quinazolinone/Amidine Rearrangement (SQuAReS).

In a round-bottom flask, compound 6 or 7 (1 equiv) was suspended in HFIP28 (0.2 M). MsOH (5 equiv) was added, and the reaction was heated for 3 h at 50 °C. After 3 h, the reaction was cooled to rt and quenched with MeOH (0.2 M). Solvent was removed under reduced pressure by azeotroping with chloroform (3×). The residue was transferred to a new sealed tube, suspended in CH3CN (0.08 M), and NEt3 (15 equiv) was added. The tube was sealed and heated to 80 °C, generally for 8 h, or until the free amine was fully converted to the cyclized product, as monitored by LCMS. Upon completion, solvent was removed under reduced pressure. The residue was then suspended in 20% IPA/CHCl3 (120 mL) and washed with brine (80 mL). The separated organic extract was dried over Na2SO4, filtered, and concentrated. The residue was then purified by normal phase chromatography to afford (±)-cis-824a, (±)-cis-26-31a, (±)-trans-824b, or (±)-trans-2630b.

General Procedure 6: N-Demethylation of Quinazolinones.

To a sealed, oven-dried pressure relief vial containing either (±)-trans-21b or (±)-trans-25b that was evacuated and backfilled with Ar (g) was added DCM (0.2 M). After the dropwise addition of ACE-Cl (4 equiv), the reaction was allowed to stir at rt for 10 min before heating to reflux for 14–16 h. After solvent and excess ACE-Cl was removed under reduced pressure, the crude reaction mixture was suspended in anhydrous methanol (0.02 M) and heated to reflux for 21 h. Upon completion, solvent was removed under reduced pressure. The crude reaction mixture was then suspended in 20% IPA/CHCl3 (250 mL) and washed with sat. aq NaHCO3 (150 mL). After drying the separated organic phase over Na2SO4 and filtering, the residue resulting from concentration was purified by normal phase chromatography to afford (±)-trans-32b or (±)-trans-33b.

General Procedure 7: N-Alkylation of NH-Quinazolinones.

In a 2 dram pressure relief vial flushed with Ar, (±)-trans-32b or 33b (1 equiv), alkyl-halide (4 equiv), and Et3N (6 equiv) were suspended in anhydrous DMF (0.25 M) and heated at 50–80 °C until starting material was consumed, as indicated by LCMS (~20 h). The reaction was diluted with 20% IPA/CHCl3 (60 mL), washed sequentially with sat. aq NaHCO3 (1 × 40 mL), H2O (2 × 40 mL) and brine (1 × 40 mL). The separated organic extract was dried over Na2SO4, filtered, and concentrated. The residue was purified by normal phase chromatography to afford (±)-trans-3441b, (±)-trans-4750b, (±)-trans-5357b, (±)-trans-5962b, and (±)-trans-65b.

General Procedure 8: N-Acylation of NH-Quinazolinones.

In a 2 dram pressure relief vial flushed with Ar, (±)-trans-32b or 33b (1 equiv) and Et3N (2 equiv) were suspended in anhydrous DCM (0.25 M) and cooled to 0 °C before an appropriately substituted acid chloride (1.2 equiv) was added dropwise. After stirring at 0 °C for 10 min, the reaction was allowed to warm to rt and then stirred until LCMS showed minimal or no starting material (~18–24 h). The reaction was diluted with 20% IPA/CHCl3 and washed with sat. aq NaHCO3. The separated organic extract was dried over Na2SO4, filtered, and concentrated to afford a residue that was purified by normal phase chromatography to afford (±)-trans-4246b.

General Procedure 9: N-Alkylation of N-Ethanolamine.

In a 2 dram pressure relief vial, (±)-trans-48b or 56b (1 equiv) and Et3N (2 equiv) were suspended in anhydrous DCE (0.25 M) and then cooled to 0 °C. At 0 °C, MsCl (1.2 equiv) was added dropwise, and then the reaction was allowed to warm to rt and stirred for 2 h. Amine (6 equiv) was added dropwise, and the reaction stirred at rt for 22 h. The reaction was diluted with 20% IPA/CHCl3 (60 mL), washed sequentially with sat. aq NaHCO3 (40 mL), water (40 mL), and brine (40 mL). The separated organic extract was dried over Na2SO4, filtered, and concentrated to afford a residue that was purified by normal phase chromatography to give (±)-trans-51–52b or (±)-trans-63–64b.

(±)-(4aR,13bR)-9-Chloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-cis-10a and (±)-(4aR,13bS)-9-Chloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-trans-10b.

Step 1: Synthesis of tert-Butyl (4-(5-Chloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)butyl)(methyl)carbamate (S1).

Compound S1 was synthesized according to general procedure 1 from 2-amino-6-chloro-benzoic acid and 2a. Purification by flash chromatography (5–25% X/hexanes, X = 4:1 EtOAc/DCM) delivered S1 as a yellow oil (1.25 g, 70%). 1H NMR (400 MHz, CDCl3) δ 7.56–7.45 (m, 2H), 7.39 (dd, J = 7.5, 1.5 Hz, 1H), 4.61–3.62 (m, 2H), 3.25 (t, J = 7.1 Hz, 2H), 2.84 (d, J = 6.1 Hz, 5H), 1.76 (t, J = 7.8 Hz, 2H), 1.68–1.57 (m, 2H), 1.44 (s, 9H), 0.99 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 161.0, 155.9, 149.4, 134.1, 133.5, 129.0, 126.1, 117.8, 79.4, 52.2, 48.7, 47.9, 35.6, 34.5, 34.2, 28.8, 28.5, 27.3, 24.5. HRMS (ESI) m/z: [M + H]+ Calcd for C23H35ClN3O3, 436.2361. Found 436.2353.

Step 2: Synthesis of 5-Chloro-2-(4-(methylamino)butyl)-3-neopentylquinazolin-4(3H)-one bis-Hydrochloride Salt (5a).

Compound 5a was synthesized according to general procedure 2 from S1, which afforded 5a as a white solid (0.90 g, 84%).

1H NMR (400 MHz, DMSO) δ 9.14 (d, J = 7.6 Hz, 2H), 7.83–7.71 (m, 2H), 7.58 (dd, J = 7.5, 1.5 Hz, 1H), 4.06 (s, 2H), 3.03 (t, J = 7.0 Hz, 2H), 2.90 (p, J = 6.7 Hz, 2H), 2.50–2.45 (m, 4H), 1.80 (dt, J = 10.3, 6.3 Hz, 4H), 0.93 (s, 9H). 13C NMR (101 MHz, DMSO) δ 160.6, 159.0, 145.4, 134.9, 132.9, 129.6, 123.2, 116.5, 51.9, 47.6, 34.0, 33.0, 32.3, 28.1, 24.7, 23.9. HRMS (ESI) m/z: [M + H]+ Calcd for C18H27ClN3O, 336.1837. Found 336.1837.

Step 3: Synthesis of Benzyl (2-(3-(5-Chloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)-1-methylpiperidin-2-yl)ethyl)-carbamate (6a).

Compound 6a was synthesized according to general procedure 3 from 5a. Purification by reverse phase flash chromatography (10–90% MeOH/H2O) which afforded 6a as an off-white solid (0.44 g, 69%). 6a was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-9-Chloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-10a and (±)-(4aR,13bS)-9-Chloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-10b.

Both diastereomers of 10 were synthesized according to general procedure 5 starting from 6a. Purified by flash chromatography10–60% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH). (±)-cis-10a and trans-10b: 138 mg (76%).

(±)-cis-10a, isolated as a light brown solid (92 mg).

1H NMR (400 MHz, CDCl3) δ 7.55–7.45 (m, 2H), 7.37 (dd, J = 6.6, 2.4 Hz, 1H), 4.07 (ddd, J = 14.2, 5.8, 3.9 Hz, 1H), 3.95 (ddd, J = 14.2, 11.2, 5.0 Hz, 1H), 2.97 (q, J = 3.6 Hz, 1H), 2.84–2.72 (m, 2H), 2.53 (dt, J = 6.0, 3.1 Hz, 1H), 2.35 (dtd, J = 14.4, 5.1, 3.9 Hz, 1H), 2.27 (s, 3H), 2.17 (td, J = 10.8, 3.3 Hz, 1H), 1.92 (dddd, J = 14.2, 11.2, 5.9, 2.7 Hz, 1H), 1.69–1.47 (m, 3H). 13C NMR (101 MHz, CDCl3) δ 160.6, 156.9, 150.0, 133.7, 133.2, 128.7, 126.3, 117.4, 57.8, 55.8, 42.9, 42.0, 39.8, 26.8, 24.8, 22.6. HRMS (ESI) m/z: [M + H]+ Calcd for C16H19ClN3O, 304.1211. Found 304.1202. HPLC Purity: ≥ 99%, RT = 1.90 min.

(±)-trans-10b, isolated as a light brown solid (46 mg).

1H NMR (400 MHz, CDCl3) δ 7.56–7.50 (m, 2H), 7.40 (dd, J = 6.4, 2.6 Hz, 1H), 4.16 (ddd, J = 14.5, 6.1, 4.8 Hz, 1H), 4.02 (ddd, J = 14.9, 9.6, 5.9 Hz, 1H), 2.96 (ddt, J = 11.7, 4.1, 1.9 Hz, 1H), 2.77–2.69 (m, 1H), 2.65 (td, J = 11.1, 3.7 Hz, 1H), 2.44 (ddt, J = 13.4, 5.9, 4.7 Hz, 1H), 2.36 (s, 3H), 2.10 (td, J = 12.0, 3.2 Hz, 1H), 1.96 (dt, J = 10.4, 5.2 Hz, 1H), 1.91–1.70 (m, 3H), 1.43 (tdd, J = 13.0, 11.5, 4.4 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.2, 156.4, 150.1, 133.9, 133.5, 129.0, 126.5, 117.5, 62.3, 56.9, 44.9, 42.7, 40.8, 27.2, 26.7, 25.2. HRMS (ESI) m/z: [M + H]+ Calcd for C16H19ClN3O, 304.1211. Found 304.1200. HPLC Purity: ≥98%, RT = 1.99 min.

(±)-(4aR,13bR)-4-Methyl-10-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Cis-15a and (±)-(4aR,13bS)-4-Methyl-10-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-15b.

Step 1: Synthesis of tert-Butyl Methyl(4-(3-neopentyl-4-oxo-6-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)butyl)carbamate (S2).

Compound S2 was synthesized according to general procedure 1 from 2-amino-5-trifluoromethyl-benzoic acid and 2a.

Purification by flash chromatography (5–20% X/hexanes, X = 4:1 EtOAc/DCM) delivered S2 as a yellow oil (1.16 g, 56%). 1H NMR (400 MHz, CDCl3) δ 8.54–8.49 (m, 1H), 7.89 (dd, J = 8.6, 2.2 Hz, 1H), 7.69 (d, J = 8.5 Hz, 1H), 4.12 (s, 2H), 3.28 (t, J = 6.9 Hz, 2H), 2.88 (d, J = 29.4 Hz, 5H), 1.79 (d, J = 9.8 Hz, 2H), 1.65 (p, J = 7.6 Hz, 2H), 1.45 (s, 9H), 1.01 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 162.5, 156.0, 149.1, 130.4, 129.7, 127.8, 125.2 (q, J = 4.2 Hz), 122.6, 120.5, 115.5, 79.5, 52.6, 47.9, 35.8, 34.8, 34.3, 28.8, 28.6, 27.4, 24.6. 19F NMR (376 MHz, CDCl3) δ − 62.31. HRMS (ESI) m/z: [M + H]+ Calcd for C24H35F3N3O3, 470.2625. Found 470.2620.

Step 2: Synthesis of 2-(4-(Methylamino)butyl)-3-neopentyl-6-(trifluoromethyl)quinazolin-4(3H)-one bis-hydrochloride Salt (5b).

Compound 5b was synthesized according to general procedure 2 from S2, which afforded 5b as a white solid (0.71 g, 80%). 1H NMR (400 MHz, DMSO) δ 8.92 (d, J = 7.9 Hz, 2H), 8.16 (d, J = 2.2 Hz, 1H), 7.92 (dd, J = 8.7, 2.2 Hz, 1H), 7.66 (d, J = 8.5 Hz, 1H), 6.02 (s, 1H), 3.93 (s, 2H), 2.81 (t, J = 7.1 Hz, 2H), 2.73 (q, J = 6.8 Hz, 2H), 2.32 (m, 3H), 1.72–1.53 (m, 4H), 0.76 (s, 9H). 13C NMR (101 MHz, DMSO) δ 161.3, 160.9, 148.0, 130.5, 127.5, 126.5 (q, J = 32.7 Hz), 124.0 (q, J = 4.1 Hz), 122.4, 119.9, 51.9, 47.8, 34.2, 34.0, 32.2, 28.3, 24.8, 23.7. 19F NMR (376 MHz, DMSO) δ − 61.6. HRMS (ESI) m/z: [M + H]+ Calcd for C19H27F3N3O, 370.2101. Found 370.3709.

Step 3: Synthesis of Benzyl (2-(1-Methyl-3-(3-neopentyl-4-oxo-6-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)piperidin-2-yl)ethyl)-carbamate (6b). Compound 6b was synthesized according to general procedure 3 from 5b.

Purification by reverse phase flash chromatography (10–90% MeOH/H2O) which afforded 6b as an off-white solid (319 mg, 76%). 6b was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-4-Methyl-10-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-15a and (±)-(4aR,13bS)-4-Methyl-10-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-15b.

Both diastereomers of 15 were synthesized according to general procedure 5 starting from 6b. Purified by flash chromatography: 5–50% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH). (±)-cis-15a and (±)-trans-15b: 128 mg (79%).

(±)-cis-15a was isolated as a tan solid (49 mg). 1H NMR (400 MHz, CDCl3) δ 8.53 (d, J = 2.1 Hz, 1H), 7.86 (dd, J = 8.6, 2.2 Hz, 1H), 7.69 (d, J = 8.6 Hz, 1H), 4.14 (ddd, J = 14.2, 5.8, 3.9 Hz, 1H), 4.02 (ddd, J = 14.0, 11.2, 5.0 Hz, 1H), 3.01 (p, J = 4.8, 4.1 Hz, 1H), 2.91–2.73 (m, 2H), 2.56 (dt, J = 5.9, 3.1 Hz, 1H), 2.37 (dq, J = 14.3, 4.8 Hz, 1H), 2.28 (s, 3H), 2.19 (td, J = 10.7, 3.8 Hz, 1H), 1.96 (dddd, J = 14.3, 11.3, 5.8, 2.7 Hz, 1H), 1.59 (dddd, J = 21.9, 11.5, 8.4, 4.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 161.2, 157.8, 149.7, 130.1 (q, J = 3.3 Hz), 128.1, 125.1, 124.6 (q, J = 4.1 Hz), 122.4, 120.0, 62.2, 56.8, 44.9, 42.6, 40.5, 27.1, 26.4, 25.0. 19F NMR (376 MHz, CDCl3) δ − 62.25. HRMS (ESI) m/z: [M + H]+ Calcd for C17H19F3N3O, 338.1375. Found 338.1464. HPLC purity: ≥99%, RT = 2.21 min.

(±)-trans-15b was isolated as a tan solid (79 mg). 1H NMR (400 MHz, CDCl3) δ 8.54 (d, J = 2.1 Hz, 1H), 7.89 (dd, J = 8.6, 2.2 Hz, 1H), 7.73 (d, J = 8.6 Hz, 1H), 4.25–4.10 (m, 2H), 3.00 (ddt, J = 11.7, 4.1, 1.8 Hz, 1H), 2.73 (qd, J = 12.1, 10.8, 4.0 Hz, 2H), 2.46 (dq, J = 13.4, 5.4 Hz, 1H), 2.39 (s, 3H), 2.14 (td, J = 12.0, 3.2 Hz, 1H), 2.01 (td, J = 10.2, 4.7 Hz, 1H), 1.94–1.74 (m, 3H), 1.56–1.43 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 161.2, 157.8, 149.7, 130.1 (q, J = 3.3 Hz), 128.1, 125.1, 124.6 (q, J = 4.1 Hz), 122.4, 120.0, 62.2, 56.8, 44.9, 42.6, 40.5, 27.1, 26.4, 25.0. 19F NMR (376 MHz, CDCl3) δ − 62.25. HRMS (ESI) m/z: [M + H]+ Calcd for C17H19F3N3O, 338.1375. Found 338.1471. HPLC purity: ≥99%, RT = 2.51 min.

(4aR,13bS)-4-Methyl-10-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-15b-ent 1 and (4aS,13bR)-4-Methyl-10-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, Trans-15b-ent 2.

60 mg of (±)-trans-15b was separated under the following conditions: Reflect I-Cellulose B5 μm (3,5-diphenylcarbamate), 25 cm × 21.1 mm. 2–15% IPA/hexanes, 1.5 mL injection, 21 mL/min.

(4aR,13bS)-4-Methyl-10-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, trans-15b-ent 1.

Absolute stereochemistry arbitrarily assigned. 22.8 mg isolated from chiral separation. (96% ee, RT = 12.93 min).

1H NMR (400 MHz, CDCl3) δ 8.53 (d, J = 2.1 Hz, 1H), 7.88 (dd, J = 8.6, 2.2 Hz, 1H), 7.72 (d, J = 8.6 Hz, 1H), 4.23–4.10 (m, 2H), 2.98 (dq, J = 11.5, 2.2 Hz, 1H), 2.79–2.66 (m, 2H), 2.45 (dq, J = 13.4, 5.4 Hz, 1H), 2.37 (s, 3H), 2.12 (td, J = 12.0, 3.1 Hz, 1H), 2.00 (td, J = 10.1, 4.7 Hz, 1H), 1.91–1.74 (m, 3H), 1.48 (qd, J = 12.6, 3.7, 1.6 Hz, 1H).13C NMR (101 MHz, CDCl3) δ 161.4, 158.0, 149.8, δ 130.28 (q, J = 3.2 Hz), 128.2, 125.3, 124.71 (q, J = 4.1 Hz), 122.6, 120.2, 62.3, 56.9, 45.0, 42.8, 40.7, 27.3, 26.6, 25.1. 19F NMR (376 MHz, CDCl3) δ − 62.25. HRMS (ESI) m/z: [M + H]+ Calcd for C17H19F3N3O, 338.1375. Found 338.1470. HPLC purity: ≥98%, RT = 2.51 min.

(4aS,13bR)-4-Methyl-10-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-15b-ent 2.

Absolute stereochemistry arbitrarily assigned. 20.6 mg isolated from chiral separation. (97% ee, RT = 15.08 min).

1H NMR (400 MHz, CDCl3) δ 8.53 (d, J = 2.2 Hz, 1H), 7.88 (dd, J = 8.7, 2.2 Hz, 1H), 7.72 (d, J = 8.6 Hz, 1H), 4.26–4.09 (m, 2H), 2.98 (dt, J = 11.9, 3.3 Hz, 1H), 2.80–2.66 (m, 2H), 2.37 (s, 4H), 2.12 (td, J = 11.9, 3.1 Hz, 1H), 2.00 (td, J = 10.2, 4.8 Hz, 1H), 1.91−1.76 (m, 3H), 1.48 (qd, J = 13.0, 12.4, 4.1 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 161.4, 158.0, 149.8, δ 130.28 (q, J = 3.3 Hz), 128.2, 125.3, 124.71 (q, J = 4.1 Hz), 122.6, 120.2, 62.3, 56.9, 45.1, 42.8, 40.7, 27.3, 26.6, 25.1. 19F NMR (376 MHz, CDCl3) δ − 62.25. HRMS (ESI) m/z: [M + H]+ Calcd for C17H19F3N3O, 338.1375. Found 338.1471. HPLC purity: ≥99%, RT = 2.51 min.

(4aR,13bS)-10-Chloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-16b-ent-1 and (4aS,13bR)-10-Chloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-16b-ent-2.

50 mg of (±)-trans-16b was separated under the following conditions: Reflect I-Cellulose B5 μm (3,5-diphenylcarbamate), 25 cm × 21.1 mm. 2–15% IPA/hexanes, 1.5 mL injection, 21 mL/min.

(4aR,13bS)-10-Chloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-16b-ent-1.

Absolute stereochemistry arbitrarily assigned. Fourteen mg isolated from chiral separation (>99% ee, RT = 9.17 min). 1H NMR (400 MHz, CDCl3) δ 8.20 (d, J = 2.4 Hz, 1H), 7.63 (dd, J = 8.8, 2.4 Hz, 1H), 7.57 (d, J = 8.7 Hz, 1H), 4.21–4.09 (m, 2H), 2.98 (ddt, J = 11.6, 4.0, 1.9 Hz, 1H), 2.75–2.65 (m, 2H), 2.47–2.40 (m, 1H), 2.37 (s, 3H), 2.11 (td, J = 12.0, 3.2 Hz, 1H), 1.99 (td, J = 10.2, 5.0 Hz, 1H), 1.89–1.73 (m, 3H), 1.53–1.41 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 161.1, 156.0, 146.2, 134.6, 132.1, 128.9, 126.0, 121.4, 62.4, 56.9, 44.9, 42.8, 40.6, 27.3, 26.6, 25.1. HRMS (ESI) m/z: [M + H]+ Calcd for C16H19ClN3O, 304.1211. Found 304.1209. HPLC purity: ≥95%, RT = 2.02 min.

(4aS,13bR)-10-Chloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-16b-ent-2.

Absolute stereochemistry arbitrarily assigned. 14.1 mg isolated from the chiral separation (>99% ee, RT = 11.03 min).

1H NMR (400 MHz, CDCl3) δ 8.21 (d, J = 2.4 Hz, 1H), 7.63 (dd, J = 8.8, 2.4 Hz, 1H), 7.57 (d, J = 8.7 Hz, 1H), 4.21–4.09 (m, 2H), 2.98 (ddt, J = 11.6, 4.1, 1.9 Hz, 1H), 2.71 (ddd, J = 12.5, 7.4, 3.8 Hz, 2H), 2.43 (dq, J = 13.3, 5.5 Hz, 1H), 2.37 (s, 3H), 2.12 (td, J = 12.0, 3.2 Hz, 1H), 1.98 (td, J = 10.3, 5.0 Hz, 1H), 1.88–1.73 (m, 3H), 1.53–1.41 (m, 1H).13C NMR (101 MHz, CDCl3) δ 161.1, 156.0, 146.2, 134.6, 132.1, 128.9, 126.0, 121.4, 62.4, 56.9, 44.8, 42.8, 40.6, 27.3, 26.6, 25.1. HRMS (ESI) m/z: [M + H]+ Calcd for C16H19ClN3O, 304.1211. Found 304.1209. HPLC purity: ≥97%, RT = 2.02 min.

(±)-(4aR,13bR)-10,11-Dichloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Cis-21a and (±)-(4aR,13bS)-10,11-Dichloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-21b.
Step 1: Synthesis of tert-Butyl (4-(6,7-Dichloro-3-neopentyl-4oxo-3,4-dihydroquinazolin-2-yl)butyl)(methyl)carbamate (S3).

Compound S3 was synthesized according to general procedure 1 from 2-amino-4,5-dichloro-benzoic acid and 2a. Purification by flash chromatography (0–25% X/hexanes, X = 4:1 EtOAc/DCM) delivered S3 as a colorless oil (0.98 g, 54%).

1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.70 (s, 1H), 4.06 (s, 2H), 3.27–3.22 (m, 2H), 2.88–2.82 (m, 5H), 1.78–1.73 (m, 2H), 1.66–1.58 (m, 2H), 1.44 (s, 9H), 0.98 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 161.7, 159.5, 155.9, 146.1, 138.7, 130.6, 128.5, 128.4, 120.2, 79.4, 52.6, 48.7, 35.6, 34.8, 34.3, 28.8, 28.6, 27.4, 24.5. HRMS (ESI) m/z: [M + H]+ Calcd for C23H34Cl2N3O3, 470.1972. Found 470.1995.

Step 2: Synthesis of 6,7-Dichloro-2-(4-(methylamino)butyl)-3-neopentylquinazolin-4(3H)-one bis-hydrochloride Salt (5c).

Compound 5c was synthesized according to general procedure 2 from S3, which afforded 5c as a white solid (0.66 g, 91%). 1H NMR (400 MHz, DMSO) δ 8.95 (s, 2H), 8.21 (s, 1H), 7.91 (s, 1H), 6.03 (s, 1H), 4.15–3.98 (m, 2H), 2.92 (dt, J = 12.4, 6.4 Hz, 4H), 2.51 (d, J = 2.0 Hz, 2H), 1.86–1.69 (m, 4H), 0.93 (s, 9H). 13C NMR (101 MHz, DMSO) δ 160.7, 159.9, 145.7, 137.2, 128.9, 128.2, 127.8, 120.0, 51.8, 47.9, 34.2, 34.1, 32.3, 28.3, 24.8, 23.5. HRMS (ESI) m/z: [M + H]+ Calcd for C18H26Cl2N3O, 370.1447. Found 370.1442.

Step 3: Synthesis of Benzyl (2-(3-(6,7-Dichloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)-1-methylpiperidin-2-yl)ethyl)-carbamate (6c).

Compound 6c was synthesized according to general procedure 3 from 5c. Purification by reverse phase flash chromatography (10–90% MeOH/H2O) which afforded 6c as an off-white solid (449 mg, 92%). 6c was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-10,11-Dichloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-21a and (±)-(4aR,13bS)-10,11-Dichloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-21b.

Both diastereomers of 21 were synthesized according to general procedure 5 starting from 6c Purified by flash chromatography: 15–50% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH). (±)-cis-21a and (±)-trans-21b: 165 mg (81%).

(±)-cis-21a, isolated as a yellow solid (60 mg). 1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.72 (s, 1H), 4.11 (ddd, J = 14.2, 5.8, 3.8 Hz, 1H), 3.97 (ddd, J = 14.1, 11.4, 5.0 Hz, 1H), 2.97 (q, J = 3.5 Hz, 1H), 2.86–2.73 (m, 2H), 2.53 (dt, J = 5.9, 3.2 Hz, 1H), 2.35 (dq, J = 14.2, 4.7 Hz, 1H), 2.27 (s, 3H), 2.17 (td, J = 10.0, 9.4, 3.4 Hz, 1H), 1.94 (dddd, J = 14.4, 11.4, 5.8, 2.8 Hz, 1H), 1.68–1.47 (m, 3H). 13C NMR (101 MHz, CDCl3) δ 161.0, 157.7, 146.7, 138.4, 130.2, 128.6, 127.8, 119.8, 58.0, 56.1, 42.9, 42.2, 39.9, 26.8, 25.1, 22.5. HRMS (ESI) m/z: [M + H]+ Calcd for C16H18Cl2N3O, 338.0821. Found 338.0809. HPLC purity: ≥97%, RT = 2.69 min.

(±)-trans-21b, isolated as a tan solid (105 mg). 1H NMR (400 MHz, CDCl3) δ 8.29 (s, 1H), 7.75 (s, 1H), 4.14 (ddt, J = 8.9, 5.9, 2.8 Hz, 2H), 3.03–2.94 (m, 1H), 2.68 (td, J = 11.5, 10.7, 3.6 Hz, 2H), 2.44 (dq, J = 13.4, 5.6 Hz, 1H), 2.37 (s, 3H), 2.12 (td, J = 12.0, 3.1 Hz, 1H), 1.99 (td, J = 10.1, 4.9 Hz, 1H), 1.94−1.70 (m, 3H), 1.45 (td, J = 13.2, 12.8, 4.0 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.5, 157.3, 146.8, 138.7, 130.6, 128.8, 127.9, 119.8, 62.2, 56.9, 44.9, 42.8, 40.6, 27.2, 26.6, 25.1. HRMS (ESI) m/z: [M + H]+ Calcd for C16H18Cl2N3O, 338.0821. Found 338.0817. HPLC purity: ≥98%, RT = 2.75 min.

(4aR,13bS)-10,11-Dichloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, trans-21b-ent-1 and (4aS,13bR)-10,11-Dichloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-21b-ent-2.

70 mg of (±)-trans-21b was separated under the following conditions: Reflect I-Cellulose B5 μm (3,5-diphenylcarbamate), 25 cm × 21.1 mm. 2–15% IPA/hexanes, 1.5 mL injection, 23 mL/min.

(4aR,13bS)-10,11-Dichloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-21b-ent-1.

Absolute stereochemistry arbitrarily assigned. Twenty-eight mg isolated from the chiral separation (>99% ee, RT = 23.63 min). 1H NMR (400 MHz, CDCl3) δ 8.26 (s, 1H), 7.73 (s, 1H), 4.12 (t, J = 6.1 Hz, 2H), 2.97 (d, J = 11.4 Hz, 1H), 2.72–2.63 (m, 2H), 2.46–2.32 (m, 4H), 2.10 (td, J = 12.0, 3.1 Hz, 1H), 1.97 (td, J = 10.0, 4.8 Hz, 1H), 1.90–1.70 (m, 3H), 1.43 (qd, J = 13.2, 12.6, 3.8 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.5, 157.3, 146.7, 138.6, 130.6, 128.8, 127.9, 119.8, 62.2, 56.9, 44.9, 42.7, 40.6, 27.1, 26.5, 25.1. HRMS (ESI) m/z: [M + H]+ Calcd for C16H18Cl2N3O, 338.0821. Found 338.0817. HPLC purity: ≥98%, RT = 2.75 min.

(4aS,13bR)-10,11-Dichloro-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-21b-ent-2.

Absolute stereochemistry arbitrarily assigned. Nineteen mg isolated from the chiral separation (>99% ee, RT = 27.42 min) 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.74 (s, 1H), 4.13 (t, J = 6.4 Hz, 2H), 2.98 (d, J = 11.6 Hz, 1H), 2.76–2.64 (m, 2H), 2.50–2.35 (m, 4H), 2.12 (td, J = 11.9, 3.2 Hz, 1H), 1.99 (td, J = 10.0, 4.9 Hz, 1H), 1.92–1.74 (m, 3H), 1.44 (qd, J = 12.9, 4.4 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.5, 157.2, 146.8, 138.7, 130.7, 128.8, 127.9, 119.8, 62.3, 56.9, 44.8, 42.7, 40.5, 27.1, 26.5, 25.0. HRMS (ESI) m/z: [M + H]+ Calcd for C16H18Cl2N3O, 338.0821. Found 338.0817. HPLC purity: ≥98%, RT = 2.75 min.

(±)-(4aR,13bR)-10-Chloro-4-methyl-11-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-22a and (±)-(4aR,13bS)-10-Chloro-4-methyl-11-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-22b.
Step 1: Synthesis of tert-Butyl (4-(6-Chloro-3-neopentyl-4-oxo-7-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)butyl)(methyl)carbamate (S4).

Compound S4 was synthesized according to general procedure 1 from 2-amino-4-chloro-5-trifluoromethyl-benzoic acid and 2a. Purification by flash chromatography (5–20% X/hexanes, X = 4:1 EtOAc/DCM) delivered S4 as an orange oil (1.34 g, 32%). 1H NMR (400 MHz, CDCl3) δ 8.54 (s, 1H), 7.72 (s, 1H), 4.36–3.92 (m, 2H), 3.27 (t, J = 6.9 Hz, 2H), 2.86 (d, J = 18.7 Hz, 5H), 1.77 (d, J = 11.5 Hz, 2H), 1.68–1.58 (m, 2H), 1.45 (s, 9H), 0.99 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 162.0, 156.0, 149.7, 137.4, 129.5, 127.6, 126.4, 124.0, 121.3, 118.7, 115.5, 79.5, 52.7, 35.8, 34.8, 34.3, 28.8, 28.6, 27.4, 24.4. 19F NMR (376 MHz, CDCl3) δ − 62.11. HRMS (ESI) m/z: [M + H]+ Calcd for C24H34ClF3N3O3, 504.2235. Found 504.2265.

Step 2: Synthesis of 6-Chloro-2-(4-(methylamino)butyl)-3-neopentyl-7-(trifluoromethyl)quinazolin-4(3H)-one bis-Hydrochloride Salt (5d).

Compound 5d was synthesized according to general procedure 2 from S4, which afforded 5d as a white solid (1.01 g, 86%). 1H NMR (400 MHz, DMSO) δ 9.19 (s, 2H), 8.36 (s, 1H), 7.92 (s, 1H), 4.08 (s, 2H), 2.93 (dt, J = 29.4, 6.4 Hz, 4H), 2.48 (s, 4H), 1.79 (td, J = 10.6, 5.4 Hz, 4H), 0.92 (s, 9H). 13C NMR (101 MHz, DMSO) δ 162.4, 160.9, 149.3, 135.6, 129.0, 126.9 (q, J = 5.4 Hz), 124.0 (q, J = 31.7 Hz), 121.2, 118.5, 66.4, 52.0, 47.8, 34.3, 32.2, 28.3, 24.8, 23.5. 19F NMR (376 MHz, DMSO) δ − 61.47. HRMS (ESI) m/z: [M + H]+ Calcd for C19H26ClF3N3O, 404.1711. Found 404.1730.

Step 3: Synthesis of Benzyl (2-(3-(6-Chloro-3-neopentyl-4-oxo-7-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)-1-methylpiperidin-2-yl)ethyl)carbamate (6d).

Compound 6d was synthesized according to general procedure 3 from 5d. Purification by reverse phase flash chromatography (10–80% MeOH/H2O) which afforded 6d as a yellow solid (872 mg, 92%). 6d was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-10-Chloro-4-methyl-11-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Cis-22a and (±)-(4aR,13bS)-10-Chloro-4-methyl-11-(trifluoromethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-22b.

Both diastereomers of 22 were synthesized according to general procedure 5 starting from 6d. Purified by flash chromatography: 15–50% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH). (±)-cis-22a and (±)-trans-22b: 411 mg (85%).

(±)-cis-22a, isolated as a yellow solid (197 mg). 1H NMR (400 MHz, CDCl3) δ 8.56 (s, 1H), 7.74 (s, 1H), 4.14 (ddd, J = 14.2, 5.8, 3.6 Hz, 1H), 3.98 (ddd, J = 14.1, 11.6, 5.0 Hz, 1H), 3.00 (q, J = 3.6 Hz, 1H), 2.91–2.82 (m, 1H), 2.82–2.74 (m, 1H), 2.54 (dt, J = 5.9, 3.1 Hz, 1H), 2.38 (dtd, J = 14.0, 5.0, 3.5 Hz, 1H), 2.27 (s, 3H), 2.21–2.12 (m, 1H), 1.95 (dddd, J = 14.4, 11.6, 5.8, 2.7 Hz, 1H), 1.58 (dddd, J = 15.4, 13.3, 7.3, 4.3 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 161.3, 160.1, 150.3, 137.0, 137.0, 129.6, 127.16 (q, J = 5.6 Hz), 125.84 (q, J = 32.1 Hz), 122.75 (q, J = 273.0 Hz)., 118.3, 57.9, 56.2, 42.9, 42.4, 40.0, 26.8, 25.1, 22.5. 19F NMR (376 MHz, CDCl3) δ − 62.02. HRMS (ESI) m/z: [M + H]+ Calcd for C19H26ClF3N3O, 404.1711. Found 404.1730. HPLC purity: ≥99%, RT = 2.73 min.

(±)-trans-22b, isolated as a yellow solid (214 mg). 1H NMR (400 MHz, CDCl3) δ 8.54 (s, 1H), 7.74 (s, 1H), 4.22–4.12 (m, 1H), 4.15–4.05 (m, 1H), 2.97 (dq, J = 11.7, 2.0 Hz, 1H), 2.69 (tt, J = 10.4, 3.8 Hz, 2H), 2.44 (dq, J = 13.3, 5.4 Hz, 1H), 2.36 (s, 3H), 2.11 (td, J = 12.0, 3.1 Hz, 1H), 1.99 (td, J = 10.0, 4.8 Hz, 1H), 1.93–1.69 (m, 3H), 1.52–1.33 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 160.8, 159.5, 150.3, 137.3, 129.8, 127.17 (q, J = 5.5 Hz), 126.14 (q, J = 32.3 Hz), 122.65 (q, J = 273.1 Hz), 118.3, 62.1, 56.8, 45.1, 42.7, 40.7, 27.1, 26.5, 25.1. 19F NMR (376 MHz, CDCl3) δ − 62.11. HRMS (ESI) m/z: [M + H]+ Calcd for C17H18ClF3N3O, 372.1066. Found 372.1099. HPLC purity: ≥97%, RT = 2.80 min.

(±)-(4aR, 13bR)-10-Chloro-4-methyl-11-phenyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-23a and (±)-(4aR,13bS)-10-Chloro-4-methyl-11-phenyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-23a.
Step 1: Synthesis of tert-Butyl (4-(7-Bromo-6-chloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)butyl)(methyl)carbamate (S5).

Compound S5 was synthesized according to general procedure 1 from 2-amino-4-bromo-5-chloro-benzoic acid and 2a. Purification by flash chromatography (0–5% EtOAc/DCM) delivered S5 as an orange oil (1.14 g, 50%). 1H NMR (400 MHz, CDCl3) δ 8.25 (s, 1H), 7.90 (s, 1H), 4.14–3.95 (m, 2H), 3.26 (t, J = 6.9 Hz, 2H), 2.85 (d, J = 10.3 Hz, 5H), 1.79–1.73 (m, 2H), 1.62 (p, J = 6.9 Hz, 2H), 1.44 (d, J = 0.7 Hz, 9H), 0.97 (d, J = 0.7 Hz, 9H). 13C NMR (101 MHz, CDCl3) δ 161.8, 159.4, 155.9, 146.0, 132.3, 132.1, 129.1, 128.0, 120.7, 79.4, 52.6, 47.9, 35.6, 34.8, 34.3, 28.8, 28.6, 27.3, 24.5. HRMS (ESI) m/z: [M + H]+ Calcd for C23H34BrClN3O3, 514.1467. Found 514.1459.

Step 2: Synthesis of 7-Bromo-6-chloro-2-(4-(methylamino)-butyl)-3-neopentylquinazolin-4(3H)-one bis-Hydrochloride Salt (5e).

Compound 5e was synthesized according to general procedure 2 from S5, which afforded 5e as a yellowish white solid (0.61 g, 67%). 1H NMR (400 MHz, DMSO) δ 9.11 (s, 2H), 8.17 (s, 1H), 8.05 (s, 1H), 6.79 (s, 1H), 4.06 (s, 2H), 2.92 (dt, J = 20.8, 6.5 Hz, 4H), 2.49 (d, J = 1.5 Hz, 3H), 1.77 (dq, J = 14.2, 7.8, 7.4 Hz, 4H), 0.92 (s, 9H). 13C NMR (101 MHz, DMSO) δ 160.7, 160.0, 145.1, 131.2, 130.9, 128.1, 127.4, 120.4, 51.9, 47.8, 34.2, 33.9, 32.2, 28.3, 24.7, 23.6. HRMS (ESI) m/z: [M + H]+ Calcd for C18H26BrClN3O, 414.0942. Found 414.0930.

Step 3a: Synthesis of Benzyl (2-(3-(7-Bromo-6-chloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)-1-methylpiperidin-2-yl)-ethyl)carbamate (6e).

Compound 6e was synthesized according to general procedure 3 from 5e. Purification by reverse phase flash chromatography (10–90% MeOH/H2O) which afforded 6e as a yellowish-white solid (611 mg, 67%). 1H NMR (400 MHz, CDCl3) δ 8.26 (d, J = 6.3 Hz, 1H), 8.14 (s, 1H), 7.94 (s, 0H), 7.40 (d, J = 7.3 Hz, 1H), 7.31 (ddt, J = 10.7, 9.1, 2.9 Hz, 4H), 6.13–5.70 (m, 0H), 5.20 (d, J = 12.4 Hz, 1H), 5.11 (d, J = 11.5 Hz, 1H), 5.07–4.93 (m, 1H), 4.39 (s, 1H), 3.80–3.52 (m, 1H), 3.30 (s, 2H), 3.15–3.04 (m, 1H), 2.98–2.90 (m, 1H), 2.85 (s, 1H), 2.83–2.66 (m, 1H), 2.52 (s, 1H), 2.45 (d, J = 13.3 Hz, 1H), 2.41 (s, 2H), 1.94 (d, J = 12.6 Hz, 1H), 1.88–1.68 (m, 2H), 1.67–1.50 (m, 1H), 1.42–1.26 (m, 0H), 0.98 (d, J = 9.1 Hz, 10H). 13C NMR (101 MHz, CDCl3) δ 161.8, 161.6, 161.6, 159.5, 156.4, 156.4, 145.6, 145.4, 137.0, 136.7, 132.7, 132.5, 132.3, 131.9, 129.3, 129.0, 128.5, 128.5, 128.4, 128.2, 128.1, 128.1, 128.0, 120.7, 120.5, 69.5, 66.6, 63.1, 60.4, 56.7, 53.9, 52.4, 51.5, 46.5, 44.6, 42.3, 40.8, 40.5, 34.4, 32.2, 31.8, 31.6, 29.8, 29.4, 28.9, 28.4, 28.2, 23.5, 22.9, 21.8. HRMS (ESI) m/z: [M + H]+ Calcd for C29H37BrClN4O3, 603.1702. Found 603.1734.

Step 3b: Synthesis of Benzyl (2-(3-(6-Chloro-3-neopentyl-4-oxo-7-phenyl-3,4-dihydroquinazolin-2-yl)-1-methylpiperidin-2-yl)-ethyl)carbamate (7).

Compound 7 was synthesized according to general procedure 4 from 6e and phenylboronic acid. Purification by flash chromatography (3–30% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH)) and scavenging afforded 7 as a tan solid (173 mg, 82%). 7 was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-10-Chloro-4-methyl-11-phenyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-23a and (±)-(4aR,13bS)-10-Chloro-4-methyl-11-phenyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-23b.

Both diastereomers of 23 were synthesized according to general procedure 5 starting from 7. Purified by flash chromatography: 10–60% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH). (±)-cis-23a and (±)-trans-23b = 89 mg (81%).

(±)-cis-23a, isolated as a waxy yellow solid (32 mg).

1H NMR (400 MHz, CDCl3) δ 8.32 (s, 1H), 7.61 (s, 1H), 7.50–7.39 (m, 5H), 4.09 (q, J = 9.8, 7.7 Hz, 2H), 3.04 (s, 1H), 2.80 (d, J = 10.7 Hz, 2H), 2.62 (t, J = 5.8 Hz, 1H), 2.42–2.18 (m, 5H), 1.99 (q, J = 10.9, 7.2 Hz, 1H), 1.61 (dd, J = 27.4, 12.9 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 161.4, 156.7, 146.3, 146.2, 138.6, 130.5, 129.7, 129.5, 128.3, 128.2, 127.3, 120.2, 58.1, 55.8, 42.8, 42.0, 39.9, 26.8, 24.7, 22.4. HRMS (ESI) m/z: [M + H]+ Calcd for C22H23ClN3O, 380.1524. Found 380.1522. HPLC Purity: ≥97%, RT = 3.07 min.

(±)-trans-23b, isolated as a tan solid (57 mg).

1H NMR (400 MHz, CDCl3) δ 8.31 (s, 1H), 7.62 (s, 1H), 7.51–7.40 (m, 4H), 7.44–7.36 (m, 1H), 4.22–4.08 (m, 2H), 3.02–2.92 (m, 1H), 2.75–2.63 (m, 2H), 2.43 (dq, J = 13.4, 5.5 Hz, 1H), 2.37 (s, 3H), 2.11 (td, J = 11.9, 3.2 Hz, 1H), 1.99 (td, J = 10.0, 4.9 Hz, 1H), 1.92−1.81(m, 2H), 1.77 (qt, J = 13.2, 3.3 Hz, 1H), 1.53–1.38 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 160.9, 156.4, 146.5, 146.2, 138.4, 130.8, 129.8, 129.4, 128.4, 128.2, 127.4, 120.2, 62.4, 56.9, 44.8, 42.7, 40.5, 27.2, 26.6, 25.1. HRMS (ESI) m/z: [M + H]+ Calcd for C22H23ClN3O, 380.1524. Found 380.1523. HPLC Purity: ≥99%, RT = 3.11 min.

(±)-(4aR,13bR)-10-Chloro-11-methoxy-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-24a and (±)-(4aR,13bS)-10-Chloro-11-methoxy-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-24b.
Step 1: Synthesis of tert-Butyl (4-(6-Chloro-7-methoxy-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)butyl)(methyl) carbamate (S6).

Compound S6 was synthesized according to general procedure 1 from 2-amino-5-chloro-4-methoxybenzoic acid and 2a. Purification by flash chromatography (3–25% X/Hexanes, X = 4:1 EtOAc/DCM) delivered S6 as a yellow oil (4.40 g, 86%). 1H NMR (400 MHz, CDCl3) δ 8.18 (s, 1H), 7.02 (s, 1H), 4.18–3.95 (m, 4H), 3.32–3.18 (m, 2H), 2.91–2.78 (m, 5H), 1.89–1.68 (m, 3H), 1.67–1.57 (m, 2H), 1.43 (s, 9H), 0.97 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 161.8, 159.7, 155.9, 147.7, 128.4, 128.2, 122.4, 114.6, 107.8, 100.0, 79.4, 56.6, 52.4, 35.8, 34.8, 34.3, 28.8, 28.6, 27.5, 24.9. HRMS (ESI) m/z: [M + H]+ Calcd for C24H37ClN3O4, 466.2467. Found 466.2469.

Step 2: Synthesis of 6-Chloro-7-methoxy-2-(4-(methylamino)-butyl)-3-neopentylquinazolin-4(3H)-one bis-Hydrochloride Salt (5f).

Compound 5f was synthesized according to general procedure 2 from S6, which afforded 5f as a white solid (3.25 g, 79%). 1H NMR (400 MHz, DMSO) δ 10.66 (s, 1H), 9.22 (q, J = 6.1 Hz, 2H), 8.07 (s, 1H), 7.42 (s, 1H), 4.09 (s, 2H), 4.02 (s, 3H), 3.04 (t, J = 7.1 Hz, 2H), 2.93 (p, J = 6.9 Hz, 2H), 2.53–2.51 (m, 3H), 1.84 (p, J = 7.3 Hz, 4H), 0.95 (s, 9H). 13C NMR (101 MHz, DMSO) δ 160.6, 160.0, 159.3, 127.4, 121.5, 113.4, 106.1, 66.4, 57.0, 52.0, 47.6, 34.2, 33.3, 32.2, 28.2, 24.8, 24.0. HRMS (ESI) m/z: [M + H]+ Calcd for C19H29ClN3O2, 366.1943. Found 366.1932.

Step 3: Synthesis of Benzyl (2-(3-(6-Chloro-7-methoxy-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)-1-methylpiperidin-2-yl)-ethyl)carbamate (6f).

Compound 6f was synthesized according to general procedure 3 from 5f Purification by normal phase flash chromatography (3–25% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH)) afforded 6f as a an off-white solid (4.73 g, 82%). 6f was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-10-Chloro-11-methoxy-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-24a) and (±)-(4aR,13bS)-10-Chloro-11-methoxy-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-24b.

Both diastereomers of 24 were synthesized according to general procedure 5 starting from 6f with the following modification: the cyclization step was carried out at 95 °C for 18 h. Purified by flash chromatography: 10–50% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH). (±)-cis-24a and trans-24b: 2.26 g (80%).

(±)-cis-24a, isolated as a light yellow-white solid (0.68 g). 1H NMR (400 MHz, CDCl3) δ 8.20 (s, 1H), 7.03 (s, 1H), 4.08 (ddd, J = 14.0, 5.9, 4.3 Hz, 1H), 3.99 (s, 4H), 2.98 (q, J = 3.7 Hz, 1H), 2.77 (dq, J = 11.8, 4.4, 3.2 Hz, 2H), 2.56 (dt, J = 6.2, 3.2 Hz, 1H), 2.33 (dq, J = 14.7, 4.9 Hz, 1H), 2.28 (s, 3H), 2.19 (td, J = 10.5, 3.6 Hz, 1H), 1.94 (dddd, J = 14.1, 10.7, 5.9, 2.8 Hz, 1H), 1.69–1.51 (m, 3H). 13C NMR (101 MHz, CDCl3) δ 161.2, 159.5, 157.2, 148.3, 127.6, 122.2, 114.3, 107.8, 58.0, 56.6, 55.7, 42.9, 42.1, 39.8, 27.0, 24.6, 22.7. HRMS (ESI) m/z: [M + H]+ Calcd for C17H21ClN3O2, 334.1312. Found 334.1313. HPLC Purity: ≥98%, RT = 2.03 min.

(±)-trans-24b was isolated as a light yellow-white solid (1.58 g). 1H NMR (400 MHz, CDCl3) δ 8.19 (s, 1H), 7.04 (s, 1H), 4.20–4.06 (m, 2H), 3.99 (s, 3H), 2.96 (dq, J = 11.7, 2.2 Hz, 1H), 2.73–2.62 (m, 2H), 2.41 (dq, J = 13.3, 5.5 Hz, 1H), 2.35 (s, 3H), 2.10 (td, J = 12.0, 3.1 Hz, 1H), 1.97 (td, J = 10.1, 4.8 Hz, 1H), 1.88–1.73 (m, 3H), 1.46 (qdd, J = 12.6, 3.5, 1.4 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.7, 159.6, 156.8, 148.4, 127.7, 122.5, 114.3, 108.0, 62.4, 56.9, 56.6, 44.9, 42.7, 40.4, 27.4, 26.6, 25.2. HRMS (ESI) m/z: [M + H]+ Calcd for C17H21ClN3O2, 334.1317. Found 334.1313. HPLC Purity: ≥99%, RT = 2.07 min.

(±)-(4aR,13bS)-11-Methoxy-4-methyl-8-oxo-2,3,4,4a,5,6,8,13b-octahydro-1H-[1,6]naphthyridino[5,6-b]quinazoline-10-carbonitrile, (±)-Trans-25b.
Steps 1–4: See Synthesis of (±)-Trans-24b.Step 5: Synthesis of (±)-(4aR,13bS)-11-Methoxy-4-methyl-8-oxo-2,3,4,4a,5,6,8,13b-octahydro-1H-[1,6]naphthyridino[5,6-b]-quinazoline-10-carbonitrile ((±)-Trans-25b).

(±)-trans-24b (1 equiv), Pd2(dba)3 (10 mol %), S-Phos (20 mol %), and Zn(CN)2 (1.2 equiv) were combined in a 2–5 mL MWI tube. The tube was sealed, evacuated, and backfilled twice with Ar (g). To the tube was added DMF/H2O (99:1, 0.11 M) and the reaction was heated under MWI 120 °C for 1 h. After cooling, the reaction mixture was concentrated and the residue was suspended in MeOH (20 mL) and absorbed to an Agilent 10 g HSCX column. After rinsing the column with an additional 80 mL of MeOH and set aside, the crude material was collected by eluting with 60 mL 7N NH3/MeOH. After concentration, the residue was purified by flash chromatography (15–60% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-25b as an off-white solid (42.7 mg, 44%).1H NMR (400 MHz, CDCl3) δ 8.44 (s, 1H), 7.03 (s, 1H), 4.22–4.03 (m, 2H), 4.01 (s, 3H), 2.98 (dq, J = 12.0, 3.1, 2.4 Hz, 1H), 2.74–2.63 (m, 2H), 2.43 (dq, J = 13.5, 5.3 Hz, 1H), 2.36 (s, 3H), 2.11 (td, J = 12.0, 3.1 Hz, 1H), 1.99 (td, J = 10.2, 4.7 Hz, 1H), 1.92–1.73 (m, 3H), 1.45 (qd, J = 12.7, 3.2 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 164.1, 160.3, 159.4, 152.4, 134.3, 115.7, 114.1, 107.5, 101.8, 62.2, 56.8, 56.7, 45.1, 42.7, 40.6, 27.3, 26.5, 25.1. HRMS (ESI) m/z: [M + H]+ Calcd for C18H21ClN4O2, 325.1659. Found 325.1655. HPLC Purity: ≥99%, RT = 2.35 min.

(±)-(4aR,13bR)-10-Chloro-12-methoxy-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-26a and (±)-(4aR,13bS)-10-Chloro-12-methoxy-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-26b.
Step 1: Synthesis of tert-Butyl (4-(6-Chloro-8-methoxy-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)butyl)(methyl)carbamate (S7).

Compound S7 was synthesized according to general procedure 1 from 2-amino-3-methoxy-4-chloro benzoic acid and 2a. Purification by flash chromatography (5–20% X/hexanes, X = 4:1 EtOAc/DCM) delivered S7 as a yellow oil (1.91 g, 83%). 1H NMR (400 MHz, CDCl3) δ 7.77 (d, J = 2.2 Hz, 1H), 7.07 (d, J = 2.3 Hz, 1H), 4.40–3.75 (m, 5H), 3.22 (s, 2H), 2.94 (dd, J = 14.2, 6.6 Hz, 2H), 2.81 (s, 3H), 1.71 (d, J = 7.7 Hz, 2H), 1.61 (q, J = 7.2 Hz, 2H), 1.42 (s, 9H), 0.96 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 162.0, 155.9, 154.9, 136.6, 132.2, 122.3, 117.8, 114.8, 79.4, 56.7, 52.7, 52.0, 36.2, 34.8, 34.3, 32.1, 28.7, 28.6, 27.3, 25.3. HRMS (ESI) m/z: [M + H]+ Calcd for C24H37ClN3O4, 466.2467. Found 466.2477.

Step 2: Synthesis of 6-Chloro-8-methoxy-2-(4-(methylamino)-butyl)-3-neopentylquinazolin-4(3H)-one bis-Hydrochloride Salt (5g).

Compound 5g was synthesized according to general procedure 2 from S7, which afforded 5g as a white solid (1.36 g, 81%). 1H NMR (400 MHz, DMSO) δ 9.26–9.16 (m, 2H), 8.17 (s, 2H), 7.57 (d, J = 2.2 Hz, 1H), 7.38 (d, J = 2.2 Hz, 1H), 4.07 (s, 2H), 3.94 (s, 3H), 2.91 (dt, J = 21.8, 6.2 Hz, 4H), 2.47 (m, 2H), 1.76 (p, J = 3.2 Hz, 4H), 0.90 (s, 9H). 13C NMR (101 MHz, DMSO) δ 160.9, 157.9, 154.7, 135.2, 131.2, 121.6, 116.3, 115.3, 56.7, 51.9, 47.7, 34.3, 34.1, 32.1, 28.3, 24.8, 23.9. HRMS (ESI) m/z: [M + H]+ Calcd for C19H29ClN3O2, 366.1942. Found 366.1932.

Step 3: Synthesis of Benzyl (2-(3-(6-Chloro-8-methoxy-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)-1-methylpiperidin-2-yl)-ethyl)carbamate (6g).

Compound 6g was synthesized according to general procedure 3 from 5g. Purification by reverse phase flash chromatography (10–90% MeOH/H2O) which afforded 6g as an off-white solid (754 mg, 85%). 6g was used without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-10-Chloro-12-methoxy-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Cis-26a and (±)-(4aR,13bS)-10-Chloro-12-methoxy-4-methyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-26b.

Both diastereomers of 26 were synthesized according to general procedure 5 starting from 6g. Purified by flash chromatography: 15–50% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH). (±)-cis-26a and (±)-trans-26b = 262 mg (79%).

(±)-cis-26a, isolated as a tan yellowish white solid (58 mg). 1H NMR (400 MHz, CDCl3) δ 7.79 (d, J = 2.2 Hz, 1H), 7.06 (d, J = 2.3 Hz, 1H), 4.11 (ddd, J = 14.2, 5.8, 3.9 Hz, 1H), 4.05–3.93 (m, 4H), 3.10 (q, J = 4.2 Hz, 1H), 2.77 (ddt, J = 19.4, 11.2, 3.3 Hz, 2H), 2.57 (p, J = 2.8 Hz, 1H), 2.37 (dq, J = 14.5, 4.8 Hz, 1H), 2.29 (s, 3H), 2.20 (dt, J = 11.4, 6.8 Hz, 1H), 1.91 (dddd, J = 13.8, 11.0, 5.7, 2.2 Hz, 1H), 1.72–1.62 (m, 1H), 1.57 (dq, J = 10.0, 5.4, 4.6 Hz, 2H). 13C NMR (101 MHz, CDCl3) δ 161.4, 155.9, 155.0, 137.4, 131.8, 121.9, 117.2, 114.8, 57.7, 56.8, 55.4, 42.9, 42.3, 40.2, 27.5, 24.1, 22.8. HRMS (ESI) m/z: [M + H]+ Calcd for C17H21ClN3O2, 334.1317. Found 334.1326. HPLC purity: ≥99%, RT = 2.06 min.

(±)-trans-26b, isolated as a yellowish white solid (204 mg). 1H NMR (400 MHz, CDCl3) δ 7.79 (d, J = 2.2 Hz, 1H), 7.07 (d, J = 2.3 Hz, 1H), 4.28 (ddd, J = 14.6, 6.0, 4.2 Hz, 1H), 3.98 (s, 4H), 2.96 (ddt, J = 11.6, 3.9, 1.9 Hz, 1H), 2.81 (ddt, J = 13.2, 4.0, 2.4 Hz, 1H), 2.73 (ddd, J = 11.5, 10.5, 3.7 Hz, 1H), 2.46 (ddt, J = 13.8, 5.8, 4.2 Hz, 1H), 2.36 (s, 3H), 2.11 (td, J = 11.9, 3.4 Hz, 1H), 1.98 (td, J = 10.4, 4.1 Hz, 1H), 1.91–1.70 (m, 3H), 1.47 (qd, J = 12.9, 4.7 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 161.0, 155.3, 155.1, 137.3, 132.2, 121.9, 117.3, 114.9, 62.5, 56.9, 56.8, 45.4, 42.7, 41.4, 27.8, 26.4, 25.2. HRMS (ESI) m/z: [M + H]+ Calcd for C17H21ClN3O2, 334.1317. Found 334.1332. HPLC purity: ≥99%, RT = 2.02 min.

(±)-(4aR,13bR)-4-Methyl-10-(trifluoromethyl)-3,4,4a,5,6,13b-hexahydro-[1,4]oxazino[2′,3′:3,4]pyrido[2,1-b]quinazolin-8(2H)-one, (±)-Cis-29a and (±)-(4aR,13bS)-4-Methyl-10-(trifluoromethyl)-3,4,4a,5,6,13b-hexahydro-[1,4]oxazino[2′,3′:3,4]pyrido[2,1-b]-quinazolin-8(2H)-one, (±)-Trans-29b.
Step 1: Synthesis of tert-Butyl Methyl(2-((3-neopentyl-4-oxo-6-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)methoxy)ethyl)carbamate (S8).

Compound S8 was synthesized according to general procedure 1 from 2-amino-5-trifluoromethyl-benzoic acid and 2b. Purification by flash chromatography (1–10% EtOAc/DCM) delivered S8 as a yellow oil (0.81 g, 39%). 1H NMR (400 MHz, CDCl3) δ 8.58–8.52 (m, 1H), 7.92 (dd, J = 8.6, 2.1 Hz, 1H), 7.77 (d, J = 8.5 Hz, 1H), 4.70 (s, 2H), 4.32–4.14 (m, 2H), 3.64 (s, 2H), 3.40 (s, 2H), 2.87 (s, 3H), 1.41 (s, 9H), 1.00 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 162.4, 155.7, 148.7, 130.5, 129.2 (q, J = 32.5 Hz), 128.4, 125.2 (q, J = 3.9 Hz), 122.4, 121.2, 119.7, 79.7, 73.1, 69.6, 52.6, 48.4, 35.8, 34.8, 28.9, 28.5. 19F NMR (376 MHz, CDCl3) δ − 62.41. HRMS (ESI) m/z: [M + H]+ Calcd for C23H33F3N3O4, 472.2418. Found 472.2425.

Step 2: Synthesis of 2-((2-(Methylamino)ethoxy)methyl)-3-neopentyl-6-(trifluoromethyl)quinazolin-4(3H)-one bis-Hydrochloride Salt (5h).

Compound 5h was synthesized according to general procedure 2 from S8, which afforded 5h as an off-white solid (0.56 g, 78%). 1H NMR (400 MHz, DMSO) δ 11.77 (s, 1H), 9.39–9.28 (m, 2H), 8.37 (d, J = 2.2 Hz, 1H), 8.16 (dd, J = 8.6, 2.2 Hz, 1H), 8.04 (d, J = 8.6 Hz, 1H), 4.82 (s, 2H), 4.08 (s, 2H), 3.90 (t, J = 5.1 Hz, 2H), 3.13 (p, J = 5.4 Hz, 2H), 2.55 (t, J = 5.4 Hz, 3H), 0.94 (s, 9H). 13C NMR (101 MHz, DMSO) δ 161.3, 157.1, 147.6, 130.8 (q, J = 3.3 Hz), 128.0, δ 127.36 (q, J = 32.5 Hz), 125.1, 124.0 (q, J = 4.2 Hz), 120.5, 70.7, 66.0, 52.0, 47.5, 34.2, 32.5, 28.3. 19F NMR (376 MHz, DMSO) δ − 61.53. HRMS (ESI) m/z: [M + H]+ Calcd for C18H26F3N3O2, 372.1893. Found 372.1887.

Step 3: Synthesis of Benzyl (2-(4-Methyl-2-(3-neopentyl-4-oxo-6-(trifluoromethyl)-3,4-dihydroquinazolin-2-yl)-morpholin-3-yl)ethyl)carbamate (6h).

Compound 6h was synthesized according to general procedure 3 from 5h. Purification by reverse phase flash chromatography (10–85% MeOH/H2O) which afforded 6h as a light yellow solid (527 mg, 84%). 6h was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-4-Methyl-10-(trifluoromethyl)-3,4,4a,5,6,13b-hexahydro-[1,4]oxazino[2′,3′:3,4]pyrido[2,1-b]-quinazolin-8(2H)-one, (±)-Cis-29a and (±)-(4aR,13bS)-4-Methyl-10-(trifluoromethyl)-3,4,4a,5,6,13b-hexahydro-[1,4]oxazino-[2′,3′:3,4]pyrido[2,1-b]quinazolin-8(2H)-one, (±)-Trans-29b.

Both diastereomers of 5x were synthesized according to general procedure 5 starting from 6h. Purified by flash chromatography: 15–50% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH). (±)-cis-29a + (±)-trans-29b: 180 mg (79%).

(±)-cis-29a, isolated as a brownish yellow solid (52 mg). 1H NMR (400 MHz, CDCl3) δ 8.55 (d, J = 2.0 Hz, 1H), 7.96–7.86 (m, 2H), 4.71 (d, J = 3.7 Hz, 1H), 4.15 (ddd, J = 14.1, 5.6, 3.9 Hz, 1H), 3.98 (ddd, J = 14.0, 11.1, 4.7 Hz, 1H), 3.91–3.75 (m, 2H), 2.81 (dt, J = 6.2, 3.0 Hz, 1H), 2.66 (dt, J = 11.8, 3.0 Hz, 1H), 2.55–2.41 (m, 2H), 2.34 (s, 3H), 1.97 (dddd, J = 14.4, 11.2, 5.6, 2.6 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 161.5, 155.7, 149.5, 130.5 (q, J = 3.3 Hz), 128.5, 125.2, 124.6 (q, J = 4.2 Hz), 122.5, 120.4, 73.8, 63.6, 56.9, 53.5, 42.4, 39.6, 21.8. 19F NMR (376 MHz, CDCl3) δ − 62.4. HRMS (ESI) m/z: [M + H]+ Calcd for C16H17F3N3O2, 340.1267. Found 340.1267. HPLC purity: ≥99%, RT = 2.19 min.

(±)-trans-29b, isolated as a yellow solid (128 mg). 1H NMR (400 MHz, CDCl3) δ 8.54 (dd, J = 2.0, 1.0 Hz, 1H), 7.96–7.88 (m, 2H), 4.35 (d, J = 10.1 Hz, 1H), 4.28–4.08 (m, 3H), 3.97 (td, J = 11.7, 2.5 Hz, 1H), 2.80 (ddd, J = 12.0, 2.5, 1.3 Hz, 1H), 2.55–2.40 (m, 2H), 2.38 (s, 3H), 2.27 (td, J = 10.1, 5.2 Hz, 1H), 1.88 (dddd, J = 13.7, 9.9, 8.8, 6.7 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.9, 154.3, 149.4, 130.6 (q, J = 3.3 Hz), 128.8, 125.1, 124.7 (q, J = 4.1 Hz), 122.4, 120.4, 76.7, 67.8, 60.7, 54.9, 42.2, 40.1, 23.7. 19F NMR (376 MHz, CDCl3) δ − 62.4. HRMS (ESI) m/z: [M + H]+ Calcd for C16H17F3N3O2, 340.1267. Found 340.1270. HPLC purity: ≥99%, RT = 2.02 min.

(±)-(4aR,13bR)-10-Chloro-4-methyl-3,4,4a,5,6,13b-hexahydro-[1,4]oxazino[2′,3′:3,4]pyrido[2,1-b]quinazolin-8(2H)-one, (±)-Cis-30a and (±)-(4aR,13bS)-10-Chloro-4-methyl-3,4,4a,5,6,13b-hexahydro-[1,4]oxazino[2′,3′:3,4]pyrido[2,1-b]quinazolin-8(2H)-one, (±)-Trans-30b.
Step 1: Synthesis of tert-Butyl (2-((6-Chloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)methoxy)ethyl)-(methyl)carbamate (S9).

Compound S9 was synthesized according to general procedure 1 from 2-amino-5-chloro-benzoic acid and 2b. Purification by flash chromatography (1–20% EtOAc/DCM) delivered S9 as a slight yellow oil (548.3 mg, 41%). 1H NMR (400 MHz, Chloroform-d) δ 8.22 (d, J = 2.3 Hz, 1H), 7.67 (dd, J = 8.7, 2.4 Hz, 1H), 7.61 (d, J = 8.7 Hz, 1H), 4.67 (s, 2H), 4.21 (s, 2H), 3.63 (s, 2H), 3.40 (s, 2H), 2.87 (s, 3H), 1.42 (s, 9H), 0.99 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 162.2, 153.8, 145.1, 134.8, 133.1, 129.1, 126.6, 122.4, 79.7, 73.1, 53.6, 52.5, 34.8, 29.0, 28.6, 27.3. HRMS (ESI) m/z: [M + H]+ Calcd for C22H33ClN3O4: 438.2154; Found: 438.2148.

Step 2: Synthesis of 6-Chloro-2-((2-(methylamino)ethoxy)-methyl)-3-neopentylquinazolin-4(3H)-one bis Hydrochloride Salt (5i).

Compound 5i was synthesized according to general procedure 2 from S9, which afforded 5i as a white solid (392.5 mg, 79%). 1H NMR (400 MHz, DMSO-d6) δ 9.23 (s, 2H), 8.09 (d, J = 2.2 Hz, 1H), 7.93–7.86 (m, 2H), 6.48 (s, 1H), 4.78 (s, 2H), 4.06 (s, 2H), 3.87 (t, J = 5.0 Hz, 2H), 3.12 (t, J = 5.6 Hz, 2H), 2.55 (t, J = 5.4 Hz, 3H), 0.94 (s, 9H). 13C NMR (101 MHz, DMSO) δ 160.8, 155.1, 143.7, 134.9, 131.8, 128.6, 125.5, 121.7, 70.7, 65.9, 51.9, 47.5, 34.2, 32.5, 28.3. HRMS (ESI) m/z: [M + H]+ Calcd for C17H24ClN3O2: 338.1630; Found: 338.1629.

Step 3: Synthesis of Benzyl (2-(2-(6-Chloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)-4-methylmorpholin-3-yl)ethyl) Carbamate (6i).

Compound 6i was synthesized according to general procedure 3 from 5i. Purification by reverse phase flash chromatography (10–75% MeOH/H2O) which afforded 6i as a white solid (356 mg, 74%). 6i was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(4aR,13bR)-10-Chloro-4-methyl-3,4,4a,5,6,13b-hexahydro-[1,4]oxazino[2′,3′:3,4]pyrido[2,1-b]-quinazolin-8(2H)-one, (±)-Cis-30a and (±)-(4aR,13bS)-10-Chloro-4-methyl-3,4,4a,5,6,13b-hexahydro-[1,4]oxazino[2′,3′:3,4]pyrido-[2,1-b]quinazolin-8(2H)-one, (±)-Trans-30b.

Both diastereomers of 30 were synthesized according to general procedure 5 starting from 6i. Purified by flash chromatography: 10–60% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH). (±)-cis-30a and (±)-trans-30b: 127.6 mg (62%).

(±)-cis-30a, isolated as an off white solid (38.5 mg). 1H NMR (400 MHz, Chloroform-d) δ 8.22 (d, J = 2.5 Hz, 1H), 7.73 (d, J = 8.7 Hz, 1H), 7.66 (dd, J = 8.7, 2.4 Hz, 1H), 4.68 (d, J = 3.7 Hz, 1H), 4.12 (ddd, J = 14.1, 5.6, 4.2 Hz, 1H), 3.98 (ddd, J = 14.1, 10.9, 4.8 Hz, 1H), 3.90–3.75 (m, 2H), 2.81 (dt, J = 6.2, 3.1 Hz, 1H), 2.66 (dt, J = 11.8, 3.2 Hz, 1H), 2.54–2.37 (m, 2H), 2.34 (s, 3H), 1.95 (dddd, J = 14.8, 10.9, 5.6, 2.7 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 161.2, 153.6, 145.9, 134.8, 132.7, 129.2, 125.9, 121.6, 73.8, 63.6, 56.9, 53.3, 42.4, 39.7, 21.5. HRMS (ESI) m/z: [M + H]+ Calcd for C15H17N3O2Cl+: 306.1004; Found: 306.1005. HPLC Purity: ≥98%, RT = 1.88 min.

(±)-trans-30b, isolated as an off white solid (89.1 mg). 1H NMR (400 MHz, Chloroform-d) δ 8.20 (d, J = 2.4 Hz, 1H), 7.77 (d, J = 8.7 Hz, 1H), 7.65 (dd, J = 8.7, 2.5 Hz, 1H), 4.32 (d, J = 10.1 Hz, 1H), 4.22 (ddd, J = 11.6, 3.5, 1.3 Hz, 1H), 4.17–4.11 (m, 2H), 3.96 (td, J = 11.7, 2.5 Hz, 1H), 2.79 (ddd, J = 12.0, 2.6, 1.3 Hz, 1H), 2.49–2.41 (m, 2H), 2.37 (s, 3H), 2.24 (td, J = 10.1, 5.2 Hz, 1H), 1.85 (dddd, J = 13.8, 10.1, 8.7, 7.0 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.6, 152.4, 145.7, 134.9, 132.9, 129.5, 126.0, 121.5, 76.7, 67.7, 60.8, 55.0, 42.2, 40.1, 23.7. HRMS (ESI) m/z: [M + H]+ Calcd for C15H17N3O2Cl+: 306.1004; Found: 306.1003. HPLC purity: ≥98%, RT = 1.64 min.

(±)-(3aR,12bR)-9-Chloro-3-methyl-2,3,3a,4,5,12b-hexahydropyrrolo[3′,2′:3,4]pyrido[2,1-b]quinazolin-7(1H)-one, (±)-Cis-31a.
Step 1: Synthesis of tert-Butyl (3-(6-Chloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)propyl)(methyl)-carbamate (S10).

Compound S10 was synthesized according to general procedure 1 from 5-chloro-2-amino-benzoic acid and 2c. Purification by flash chromatography (5–20% X/hexanes, X = 4:1 EtOAc/DCM) delivered S10 as a yellow oil (4.36 g, 88%). 1H NMR (400 MHz, CDCl3) δ 8.18 (d, J = 2.4 Hz, 1H), 7.62 (dd, J = 8.7, 2.4 Hz, 1H), 7.54 (d, J = 8.7 Hz, 1H), 4.06 (s, 2H), 3.36 (t, J = 6.8 Hz, 2H), 2.91–2.80 (m, 5H), 2.01 (p, J = 7.1 Hz, 2H), 1.44 (s, 9H), 0.98 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 162.2, 145.5, 134.6, 132.1, 128.5, 126.5, 121.8, 79.6, 52.6, 48.4, 47.9, 34.8, 34.3, 32.9, 28.8, 28.6, 27.5, 25.5. HRMS (ESI) m/z: [M + H]+ Calcd for C22H33ClN3O3, 422.2205. Found 422.2199.

Step 2: Synthesis of 6-Chloro-2-(3-(methylamino)propyl)-3-neopentylquinazolin-4(3H)-one bis-Hydrochloride Salt (5j).

Compound 5j was synthesized according to general procedure 2 from S10, which afforded 5j as a white solid (3.44 g, 86%). 1H NMR (400 MHz, DMSO) δ 10.26 (s, 2H), 9.38 (q, J = 6.1 Hz, 2H), 8.03 (d, J = 2.5 Hz, 1H), 7.86 (dd, J = 8.7, 2.5 Hz, 1H), 7.75 (d, J = 8.7 Hz, 1H), 4.08 (s, 2H), 3.12 (t, J = 7.6 Hz, 2H), 3.07–2.96 (m, 2H), 2.50 (t, J = 5.6 Hz, 3H), 2.14 (p, J = 7.4 Hz, 2H), 0.92 (s, 9H). 13C NMR (101 MHz, DMSO) δ 160.6, 159.1, 142.7, 134.9, 131.2, 127.0, 125.6, 121.1, 52.1, 47.4, 34.3, 32.2, 31.4, 28.2, 23.2. HRMS (ESI) m/z: [M + H]+ Calcd for C17H25N3O, 322.1681. Found 322.1673.

Step 3: Synthesis of Benzyl (2-(3-(6-Chloro-3-neopentyl-4-oxo-3,4-dihydroquinazolin-2-yl)-1-methylpyrrolidin-2-yl)ethyl)-carbamate (6j).

Compound 6j was synthesized according to general procedure 3 from 5j. Purification by normal phase flash chromatography (3–15% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH)) afforded 6j as a yellow solid (1.63 g, 63%). 6j was used directly in the next step without analysis.

Step 4: Synthesis of (±)-(3aR,12bR)-9-Chloro-3-methyl-2,3,3a,4,5,12b-hexahydropyrrolo[3′,2′:3,4]pyrido[2,1-b]quinazolin-7(1H)-one, (±)-Cis-31a.

(±)-cis-31a was synthesized according to general procedure 5 starting from 6j with the following modification: instead of heating at 80 °C for 8h, the reaction was heated to 150 °C for 4 h. Purified by flash chromatography: 3–20% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH). (±)-cis-31a was isolated as a yellow solid (186 mg, 55%). 1H NMR (400 MHz, CDCl3) δ 8.20 (d, J = 2.4 Hz, 1H), 7.62 (dd, J = 8.7, 2.4 Hz, 1H), 7.53 (d, J = 8.7 Hz, 1H), 4.50 (dddd, J = 13.7, 4.9, 3.5, 1.1 Hz, 1H), 3.87 (ddd, J = 13.8, 11.1, 2.8 Hz, 1H), 3.54 (q, J = 9.1 Hz, 1H), 3.11–3.06 (m, 1H), 2.76 (dt, J = 8.6, 3.8 Hz, 1H), 2.42–2.32 (m, 5H), 2.04–1.90 (m, 2H), 1.79 (tt, J = 10.6, 4.0 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.9, 157.7, 146.3, 134.6, 131.9, 128.5, 126.2, 121.2, 61.7, 55.9, 44.6, 40.1, 37.3, 31.8, 27.0. HRMS (ESI) m/z: [M + H]+ Calcd for C15H17N3O, 290.1040. Found 290.1047. HPLC purity: ≥98%, RT = 1.60 min.

(±)-(4aR,13bS)-10,11-Dichloro-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one ((±)-Trans-32b). Steps 1–4: Synthesis of (±)-Trans-21b
Step 5: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-32b.

Compound (±)-trans-32b was synthesized according to general procedure 6 from (±)-trans-21b. Purification by flash chromatography: 40–75% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-32b as a light-yellow solid (134 mg, 70%). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.74 (s, 1H), 4.18 (ddd, J = 14.4, 6.3, 4.7 Hz, 1H), 4.08 (ddd, J = 14.7, 9.5, 5.9 Hz, 1H), 3.15 (ddt, J = 11.9, 4.0, 1.9 Hz, 1H), 2.70 (ddt, J = 12.1, 9.1, 4.3 Hz, 3H), 2.47 (td, J = 11.0, 3.6 Hz, 1H), 2.19 (dq, J = 13.6, 5.2 Hz, 1H), 1.85 (ddt, J = 16.8, 9.6, 3.2 Hz, 2H), 1.73–1.57 (m, 2H), 1.47 (qd, J = 13.2, 3.9 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.7, 157.2, 146.7, 138.7, 130.6, 128.8, 127.9, 119.9, 55.3, 46.6, 45.9, 41.2, 29.3, 27.3, 26.2. HRMS (ESI) m/z: [M + H]+ Calcd for C15H16Cl2N3O, 324.0665. Found 324.0672. HPLC purity: ≥99%, RT = 2.71 min.

(±)-(4aR,13bS)-10-Chloro-11-methoxy-1,2,3,4,4a,5,6,13b-octa-hydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one ((±)-Trans-33b).
Steps 1–4:

See synthesis of (±)-Trans-25b

Step 5: Synthesis of (±)-(4aR,13bS)-10-Chloro-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-33b.

Compound (±)-trans-33b was synthesized according to general procedure 6 from (±)-trans-25b. Purification by flash chromatography 55–80% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-33b as a tan solid (583 mg, 61%). 1H NMR (400 MHz, CDCl3) δ 8.20 (s, 1H), 7.04 (s, 1H), 4.18 (ddd, J = 14.3, 6.2, 4.8 Hz, 1H), 4.07 (ddd, J = 14.2, 9.6, 5.8 Hz, 1H), 3.99 (s, 3H), 3.15 (ddt, J = 11.9, 3.9, 1.8 Hz, 1H), 2.76–2.65 (m, 3H), 2.47 (td, J = 11.0, 3.6 Hz, 1H), 2.24–2.14 (m, 1H), 1.91–1.72 (m, 3H), 1.64 (qt, J = 12.9, 4.0 Hz, 1H), 1.49 (tdd, J = 13.2, 11.3, 3.8 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.9, 159.6, 156.7, 148.3, 127.7, 122.5, 114.4, 108.0, 56.7, 55.4, 46.6, 45.8, 40.9, 29.4, 27.5, 26.3. HRMS (ESI) m/z: [M + H]+ Calcd for C16H19ClN3O2, 320.1160. Found 320.1159. HPLC purity: ≥99%, RT = 2.14 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-isopropyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-34b. Steps 1–5: See synthesis of (±)-Trans-32b
Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-isopropyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-34b.

Compound (±)-trans-34b was prepared by suspending (±)-trans-32b (1 equiv), isopropyl iodide (9.25 equiv), and Et3N (4 equiv) in DMF (0.25 M) and heating to 80 °C for 20 h. Purification by flash chromatography 1–20% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-34b as a light-yellow solid (27 mg, 48%). 1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.74 (s, 1H), 4.16–4.10 (m, 2H), 3.40 (q, J = 6.6 Hz, 1H), 3.02–2.97 (m, 1H), 2.72–2.64 (m, 2H), 2.48–2.39 (m, 2H), 2.11–2.05 (m, 1H), 1.93–1.78 (m, 2H), 1.68–1.58 (m, 1H), 1.47–1.37 (m, 1H), 1.17 (d, J = 6.7 Hz, 3H), 0.88 (d, J = 6.5 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 160.5, 157.9, 146.8, 138.6, 130.5, 128.8, 127.9, 119.8, 57.6, 47.2, 45.4, 43.4, 40.6, 27.7, 25.8, 25.3, 21.9, 12.4. HRMS (ESI) m/z: [M + H]+ Calcd for C18H22Cl2N3O, 366.1134. Found 366.1152. HPLC purity: ≥99%, RT = 2.78 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-isobutyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-35b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-isobutyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-35b.

Compound (±)-trans-35b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), isobutyl bromide (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 80 °C for 18 h. Purification by flash chromatography 5–25% X/hexanes (X = 4:1 EtOAc/DCM) afforded (±)-trans-35b as a yellowish white solid (40 mg, 50%). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.75 (s, 1H), 4.20 (dt, J = 14.7, 7.1 Hz, 1H), 4.04 (dt, J = 14.4, 6.2 Hz, 1H), 3.07 (d, J = 11.4 Hz, 1H), 2.73–2.56 (m, 2H), 2.54–2.33 (m, 2H), 2.15 (q, J = 8.9 Hz, 1H), 2.03–1.87 (m, 2H), 1.75 (d, J = 37.4 Hz, 4H), 1.45 (qd, J = 13.0, 4.0 Hz, 1H), 0.92 (dd, J = 11.0, 6.5 Hz, 6H). 13C NMR (101 MHz, CDCl3) δ 160.5, 157.9, 146.9, 138.6, 130.6, 128.9, 127.9, 119.9, 61.8, 60.7, 53.7, 45.0, 40.3, 27.3, 27.2, 26.9, 24.9, 21.4, 20.9. HRMS (ESI) m/z: [M + H]+ Calcd for C19H24Cl2N4O, 380.1272. Found 380.1277. HPLC purity: ≥99%, RT = 2.96 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(cyclopropylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-36b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(cyclopropylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-36b.

Compound (±)-trans-36b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), (bromomethyl)-cyclopropane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 80 °C for 18 h. Purification by flash chromatography 0.5–6% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-36b as a light-yellow solid (52.3 mg, 65%). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.75 (s, 1H), 4.20 (ddd, J = 14.5, 8.3, 6.2 Hz, 1H), 4.06 (dt, J = 14.4, 6.1 Hz, 1H), 3.30 (ddt, J = 11.9, 4.0, 1.8 Hz, 1H), 2.77 (dd, J = 13.4, 6.1 Hz, 1H), 2.74–2.62 (m, 2H), 2.43 (dq, J = 13.4, 6.0 Hz, 1H), 2.35–2.19 (m, 3H), 1.91–1.68 (m, 3H), 1.46 (tdd, J = 12.9, 11.2, 3.8 Hz, 1H), 0.93–0.82 (m, 1H), 0.68–0.48 (m, 2H), 0.18–0.09 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 160.5, 157.6, 146.8, 138.6, 130.6, 128.9, 127.9, 119.9, 59.5, 58.3, 53.1, 44.9, 40.3, 27.1, 26.7, 25.0, 7.4, 5.0, 3.5. HRMS (ESI) m/z: [M + H]+ Calcd for C19H22Cl2N3O, 378.1134. Found 378.1140. HPLC purity: ≥99%, RT = 2.75 min.

(±)-(4aR,13bS)-4-Benzyl-10,11-dichloro-1,2,3,4,4a,5,6,13b-octa-hydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-37b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-4-Benzyl-10,11-dichloro-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-37b.

Compound (±)-trans-37b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), benzyl bromide (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 18 h. Purification by flash chromatography 0.5–8% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-37b as a white solid (74.5 mg, 74%). HPLC Purity: ≥ 99%. 1H NMR (400 MHz, CDCl3) δ 8.30 (s, 1H), 7.76 (s, 1H), 7.33 (d, J = 4.4 Hz, 4H), 7.30–7.24 (m, 1H), 4.29 (ddd, J = 14.4, 8.0, 6.3 Hz, 1H), 4.17–4.04 (m, 2H), 3.24 (d, J = 13.7 Hz, 1H), 2.95 (dq, J = 13.3, 3.5, 2.7 Hz, 1H), 2.77 (td, J = 11.1, 3.7 Hz, 1H), 2.67–2.60 (m, 1H), 2.55 (dq, J = 13.3, 6.1 Hz, 1H), 2.30 (ddd, J = 10.6, 9.1, 5.4 Hz, 1H), 2.05–1.91 (m, 2H), 1.82–1.75 (m, 1H), 1.66 (qt, J = 12.9, 3.7 Hz, 1H), 1.47 (tdd, J = 13.2, 11.5, 4.1 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.7, 146.8, 138.7, 138.6, 130.6, 129.0, 128.9, 128.5, 127.9, 127.2, 119.9, 60.2, 58.0, 53.1, 45.0, 40.1, 27.3, 27.1, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C22H22Cl2N3O, 414.1134. Found 414.1136. HPLC purity: ≥99%, RT = 2.92 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(pyridin-2-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-38b.
Steps 1–5:

See synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(pyridin-2-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-38b.

Compound (±)-trans-38b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 2-(chloromethyl)-pyridine hydrochloride (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 18 h. Purification by flash chromatography 3–30% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-38b as an off white solid (72.3 mg, 81%). 1H NMR (400 MHz, CDCl3) δ 8.55 (dd, J = 5.0, 1.7 Hz, 1H), 8.27 (s, 1H), 7.73 (s, 1H), 7.65 (td, J = 7.7, 1.8 Hz, 1H), 7.41 (d, J = 7.8 Hz, 1H), 7.20–7.12 (m, 1H), 4.25–4.03 (m, 3H), 3.56 (d, J = 14.4 Hz, 1H), 2.95 (dt, J = 11.6, 3.4 Hz, 1H), 2.76 (td, J = 11.2, 3.7 Hz, 1H), 2.70–2.54 (m, 2H), 2.37 (td, J = 10.0, 5.1 Hz, 1H), 2.18 (td, J = 11.8, 3.2 Hz, 1H), 1.97–1.64 (m, 3H), 1.45 (qd, J = 12.9, 4.4 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 158.9, 157.5, 149.4, 146.8, 138.6, 136.5, 130.6, 128.8, 127.9, 123.3, 122.2, 119.8, 60.2, 59.8, 53.7, 45.0, 40.4, 27.1, 27.1, 24.9. HRMS (ESI) m/z: [M + H]+ Calcd for C21H21Cl2N4O, 415.1087. Found 415.1090. HPLC purity: ≥99%, RT = 2.96 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(pyridin-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-39b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(pyridin-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-39b.

Compound (±)-trans-39b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 3-(bromomethyl)-pyridine hydrobromide (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 20 h. Purification by flash chromatography 5–40% X/Hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-39b as a tan solid (28.1 mg, 31%). 1H NMR (400 MHz, CDCl3) δ 8.57 (d, J = 2.2 Hz, 1H), 8.52 (dd, J = 4.8, 1.6 Hz, 1H), 8.30 (s, 1H), 7.76 (s, 1H), 7.68 (dt, J = 7.8, 2.0 Hz, 1H), 7.27 (dd, J = 7.8, 4.8 Hz, 1H), 4.32–4.24 (m, 1H), 4.17–4.05 (m, 2H), 3.22 (d, J = 14.0 Hz, 1H), 2.89 (dt, J = 11.6, 3.5 Hz, 1H), 2.76 (td, J = 11.1, 3.7 Hz, 1H), 2.68–2.61 (m, 1H), 2.58–2.48 (m, 1H), 2.33 (ddd, J = 10.7, 9.1, 5.4 Hz, 1H), 2.07–1.90 (m, 2H), 1.79 (dt, J = 13.3, 3.4 Hz, 1H), 1.66 (dddd, J = 16.7, 13.0, 8.4, 3.8 Hz, 1H), 1.48 (qd, J = 13.1, 3.9 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.4, 150.3, 148.8, 146.8, 138.7, 136.4, 134.2, 130.7, 128.9, 127.9, 123.5, 119.9, 60.4, 55.3, 53.2, 44.9, 40.0, 27.3, 27.0, 24.7. HRMS (ESI) m/z: [M + H]+ Calcd for C21H21Cl2N4O, 415.1087. Found 415.1100. HPLC purity: ≥99%, RT = 2.90 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(pyridin-4-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-40b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(pyridin-4-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one ((±)-Trans-40b).

Compound (±)-trans-40b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 2–4-(bromomethyl)-pyridine hydrobromide (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 20 h. Purification by flash chromatography 5–30% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-40b as a tan solid (38 mg, 42%). 1H NMR (400 MHz, CDCl3) δ 8.57 (d, J = 5.1 Hz, 2H), 8.30 (s, 1H), 7.77 (s, 1H), 7.35–7.30 (m, 2H), 4.25 (ddd, J = 14.5, 8.3, 6.2 Hz, 1H), 4.15–4.06 (m, 2H), 3.22 (d, J = 14.7 Hz, 1H), 2.89 (dq, J = 11.7, 2.1 Hz, 1H), 2.81–2.75 (m, 1H), 2.71–2.65 (m, 1H), 2.44 (dt, J = 13.2, 5.8 Hz, 1H), 2.35 (ddd, J = 10.6, 9.0, 5.3 Hz, 1H), 2.07 (td, J = 11.9, 2.9 Hz, 1H), 1.94–1.86 (m, 1H), 1.85–1.78 (m, 1H), 1.70 (qt, J = 13.0, 3.7 Hz, 1H), 1.50 (tdd, J = 13.2, 11.5, 4.1 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.3, 150.0, 148.6, 146.8, 138.7, 130.7, 128.9, 127.9, 123.6, 119.9, 60.5, 57.0, 53.8, 45.0, 40.1, 27.3, 27.0, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C21H21Cl2N4O, 415.1087. Found 415.1071. HPLC purity: ≥ 99%, 3.48 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(pyrazin-2-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-41b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(pyrazin-2-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-41b.

Compound (±)-trans-41b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 2-(bromomethyl)-pyrazine hydrobromide (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 20 h. Purification by flash chromatography 1–25% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-41b as an off-white solid (75.7 mg, 84%).1H NMR (400 MHz, CDCl3) δ 8.69 (d, J = 1.5 Hz, 1H), 8.53 (dd, J = 2.6, 1.5 Hz, 1H), 8.47 (d, J = 2.5 Hz, 1H), 8.28 (s, 1H), 7.74 (s, 1H), 4.26–4.07 (m, 3H), 3.64 (d, J = 14.8 Hz, 1H), 2.94 (ddt, J = 11.4, 4.0, 2.0 Hz, 1H), 2.77 (td, J = 11.2, 3.7 Hz, 1H), 2.70–2.57 (m, 2H), 2.40 (ddd, J = 10.7, 9.4, 5.1 Hz, 1H), 2.21 (td, J = 11.9, 3.1 Hz, 1H), 1.92 (dtd, J = 13.5, 8.9, 6.3 Hz, 1H), 1.84–1.68 (m, 2H), 1.51–1.39 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 160.5, 157.3, 154.4, 146.8, 145.2, 144.2, 143.4, 138.7, 130.7, 128.9, 127.9, 119.8, 60.0, 57.3, 53.8, 45.0, 40.3, 27.1, 27.1, 24.9. HRMS (ESI) m/z: [M + H]+ Calcd for C20H20Cl2N5O, 416.1039. Found 416.1040. HPLC purity: ≥99%, RT = 2.97 min.

(±)-(4aR,13bS)-4-Benzoyl-10,11-dichloro-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-42b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-4-Benzoyl-10,11-dichloro-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one ((±)-Trans-42b).

Compound (±)-trans-34b was prepared according to general procedure 8 from (±)-trans-32b (1 equiv), benzoyl chloride (2.6 equiv), Et3N (2 equiv), and DMAP (0.9 equiv) in DCM (0.25 M) with stirring at rt for 48 h. Purification by flash chromatography (5–55% EtOAc/hexanes) afforded (±)-trans-42b as a white solid (44.5 mg, 48%). 1H NMR (400 MHz, chloroform-d) δ 8.33 (s, 1H), 7.78 (s, 1H), 7.56–7.41 (m, 5H), 4.70 (dt, J = 13.9, 5.3 Hz, 1H), 4.24 (ddd, J = 13.8, 9.0, 4.5 Hz, 1H), 3.84 (dt, J = 13.7, 4.2 Hz, 1H), 3.48 (ddd, J = 11.1, 8.1, 6.0 Hz, 1H), 3.25 (ddd, J = 13.7, 10.8, 2.8 Hz, 2H), 2.71−2.53 (m, 2H), 2.36 (dtd, J = 14.0, 8.6, 5.1 Hz, 1H), 1.87–1.71 (m, 2H), 1.64–1.54 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 173.9, 160.1, 157.2, 146.7, 138.7, 136.3, 130.8, 130.8, 128.9, 128.8, 128.2, 128.0, 120.1, 58.1, 50.6, 40.7, 40.4, 26.6, 26.3, 25.9. HRMS (ESI) m/z: [M + H]+ Calcd for C22H20Cl2N3O2+: 428.0927; Found: 428.0920. HPLC purity: ≥99%, RT = 3.93 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-pivaloyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-43b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-pivaloyl-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-43b.

Compound (±)-trans-43b was prepared according to general procedure 8 from (±)-trans-32b (1 equiv), pivaloyl chloride (1.3 equiv) and Et3N (2 equiv) in DCM (0.25 M). Purification by flash chromatography 5–50% EtOAc/Hexanes afforded (±)-trans-43b as a white solid (77 mg, 87%). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.73 (s, 1H), 4.65 (dt, J = 13.9, 5.4 Hz, 1H), 4.09 (ddd, J = 13.9, 8.3, 5.4 Hz, 1H), 3.99–3.88 (m, 1H), 3.47–3.31 (m, 2H), 3.08 (td, J = 11.3, 3.5 Hz, 1H), 2.67–2.55 (m, 1H), 2.28–2.14 (m, 2H), 2.00–1.89 (m, 1H), 1.80–1.64 (m, 2H), 1.29 (s, 9H). 13C NMR (101 MHz, CDCl3) δ 180.2, 160.1, 157.5, 146.6, 138.5, 130.7, 128.8, 128.1, 120.1, 58.2, 46.8, 40.4, 40.1, 39.9, 28.5, 26.8, 25.4, 25.0. HRMS (ESI) m/z: [M + H]+ Calcd for C20H24Cl2N3O2, 408.1240. Found 408.1052. HPLC purity: ≥ 98%, RT = 3.98 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(tetrahydro-2H-pyran-4-carbonyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-44b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(tetrahydro-2H-pyran-4-carbonyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-44b.

Compound (±)-trans-44b was prepared according to general procedure 8 from (±)-trans-32b (1 equiv), tetrahydro-2H-pyran-4-carbonyl chloride (1.3 equiv), and Et3N (2 equiv) in DCM (0.25 M) and stirring at room temperature for 24h. Purification by flash chromatography 50–100% EtOAc/hexanes afforded (±)-trans-44b as an off-white solid (78 mg, 90%). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.73 (s, 1H), 4.54 (dt, J = 14.1, 6.0 Hz, 1H), 4.12 (dt, J = 13.6, 6.5 Hz, 1H), 4.02 (dtd, J = 11.2, 4.3, 2.3 Hz, 2H), 3.75 (dt, J = 14.2, 4.8 Hz, 1H), 3.52–3.36 (m, 4H), 3.02 (td, J = 11.4, 3.8 Hz, 1H), 2.78–2.63 (m, 2H), 2.35 (q, J = 6.6 Hz, 2H), 2.02–1.85 (m, 3H), 1.78–1.60 (m, 4H). 13C NMR (101 MHz, CDCl3) δ 175.7, 160.1, 156.9, 146.4, 138.5, 130.7, 128.7, 128.0, 119.9, 67.3, 67.2, 57.1, 45.5, 40.7, 40.5, 39.2, 29.6, 29.0, 26.7, 25.3, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C21H24Cl2N3O3, 436.1189. Found 436.1184. HPLC purity: ≥99%, RT = 3.65 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(morpholine-4-carbonyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-45b).
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)- (4aR,13bS)-10,11-Dichloro-4-(morpholine-4-carbonyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-45b.

Compound (±)-trans-45b was prepared according to general procedure 8 from (±)-trans-32b (1 equiv), morpholine-4-carbonyl chloride (2.6 equiv), and DMAP (0.25 equiv) in DCM (0.25 M) and heating to 40 °C for 16 h. Purification by flash chromatography 5–40% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-45b as an off-white solid (87 mg, 93%).). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.74 (s, 1H), 4.49 (dt, J = 14.0, 5.9 Hz, 1H), 4.02 (ddd, J = 13.8, 8.3, 5.1 Hz, 1H), 3.68–3.65 (m, 4H), 3.47 (q, J = 4.5 Hz, 3H), 3.39–3.33 (m, 1H), 3.27–3.24 (m, 1H), 3.12–2.98 (m, 2H), 2.82 (td, J = 12.5, 2.8 Hz, 1H), 2.68−2.59 (m, 1H), 2.37 (dddd, J = 13.9, 8.2, 6.9, 5.6 Hz, 1H), 1.93–1.82 (m, 2H), 1.71–1.57 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 163.5, 160.1, 157.4, 146.6, 138.5, 130.5, 128.7, 127.9, 119.9, 66.8, 66.6, 56.4, 51.6, 47.2, 45.8, 41.9, 40.0, 27.3, 26.7, 25.0. HRMS (ESI) m/z: [M + H]+ Calcd for C20H23Cl2N4O3, 437.1120. Found 437.1132. HPLC purity: ≥ 99%, RT = 3.80 min.

(±)- (4aR,13bS)-10,11-Dichloro-4-(dimethylglycyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-46b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)- (4aR,13bS)-10,11-Dichloro-4-(dimethyl-glycyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-46b.

Compound (±)-trans-46b was prepared according to general procedure 8 from (±)-trans-32b (1 equiv), dimethylglycinoyl chloride (1.3 equiv), and Et3N (2 equiv) in DCM (0.25 M) and stirring at rt for 23 h. Purification by flash chromatography 5–50% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-46b as an off-white solid (73 mg, 83%). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.72 (s, 1H), 4.50 (dt, J = 14.2, 6.1 Hz, 1H), 4.12 (ddd, J = 14.1, 7.3, 5.6 Hz, 1H), 3.81 (dt, J = 14.3, 5.4 Hz, 1H), 3.45 (ddt, J = 21.9, 7.7, 6.0 Hz, 2H), 3.17–2.95 (m, 3H), 2.65 (ddt, J = 8.8, 5.8, 3.2 Hz, 1H), 2.41 (dq, J = 7.4, 6.1 Hz, 2H), 2.29 (s, 6H), 2.03–1.91 (m, 1H), 1.75–1.63 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 171.4, 160.2, 157.1, 146.5, 138.6, 130.8, 128.8, 128.1, 120.0, 63.7, 57.3, 45.8, 45.6, 41.1, 40.7, 26.6, 24.9, 24.9. HRMS (ESI) m/z: [M + H]+ Calcd for C19H23Cl2N4O3, 409.1193. Found 409.1188. HPLC purity: ≥97%, RT = 2.94 min.

(±)- 2-((4aR,13bS)-10,11-Dichloro-8-oxo-2,3,5,6,8,13b-hexahydro-1H-[1,6]naphthyridino[5,6-b]quinazolin-4(4aH)-yl)-N,N-dimethylacetamide ((±)-Trans-47b).
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)- 2-((4aR,13bS)-10,11-Dichloro-8-oxo-2,3,5,6,8,13b-hexahydro-1H-[1,6]naphthyridino[5,6-b]quinazolin-4(4aH)-yl)-N,N-dimethylacetamide ((±)-Trans-47b).

Compound (±)-trans-47b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 2-bromo-N,N-dimethylacetamide (1.4 equiv), and Et3N (2 equiv) in DMF (0.25 M) and heating to 50 °C for 24 h. Purification by flash chromatography 10–50% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-47b as a tan solid (79 mg, 90%). 1H NMR (400 MHz, CDCl3) δ 8.26 (s, 1H), 7.72 (s, 1H), 4.22–4.16 (m, 1H), 4.00 (ddd, J = 14.5, 9.8, 5.9 Hz, 1H), 3.63 (d, J = 15.2 Hz, 1H), 3.27 (d, J = 15.2 Hz, 1H), 3.07 (s, 3H), 2.94 (s, 4H), 2.73–2.61 (m, 3H), 2.50 (td, J = 11.9, 3.0 Hz, 1H), 2.45–2.38 (m, 1H), 1.86–1.67 (m, 3H), 1.44 (tdd, J = 13.6, 11.0, 4.5 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 170.2, 160.5, 157.3, 146.8, 138.6, 130.5, 128.8, 127.8, 119.8, 59.4, 55.4, 53.6, 45.4, 40.9, 37.1, 35.7, 27.4, 26.3, 25.2. HRMS (ESI) m/z: [M + H]+ Calcd for C19H23Cl2N4O2, 409.1193. Found 409.1221. HPLC purity: ≥99%, RT = 2.75 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(2-hydroxyethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one ((±)-Trans-48b).
Steps 1–5:

See synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(2-hydroxyethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-48b.

Compound (±)-trans-48b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 2-bromoethanol (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 24 h. Purification by flash chromatography 10–60% X/Hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-48b as a yellow solid (54 mg, 68%). 1H NMR (400 MHz, CDCl3) δ 8.29 (s, 1H), 7.76 (s, 1H), 4.29–4.14 (m, 1H), 4.07 (dt, J = 14.0, 6.0 Hz, 1H), 3.75 (ddd, J = 12.4, 9.6, 3.7 Hz, 1H), 3.66–3.57 (m, 1H), 3.20–3.06 (m, 2H), 2.74–2.63 (m, 2H), 2.54–2.29 (m, 4H), 2.17 (td, J = 12.2, 2.7 Hz, 1H), 1.93–1.80 (m, 2H), 1.77–1.67 (m, 1H), 1.59–1.43 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.2, 146.7, 138.7, 130.7, 128.9, 127.9, 119.8, 60.3, 58.7, 54.2, 53.0, 45.0, 40.1, 27.2, 27.0, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C17H20Cl2N3O2, 368.0909. Found 368.0930. HPLC purity: ≥99%, RT = 2.75 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(2-methoxyethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-49b.
Steps 1–5:

See Synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(2-methoxyethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-49b.

Compound (±)-trans-49b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 1-bromo-2-methoxyethane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 22 h. Purification by flash chromatography 3–20% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-49b as a yellow solid (62 mg, 75%). 1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.74 (s, 1H), 4.20 (ddd, J = 14.4, 8.2, 6.2 Hz, 1H), 4.05 (dt, J = 14.4, 6.2 Hz, 1H), 3.52 (dd, J = 6.2, 5.1 Hz, 2H), 3.35 (s, 3H), 3.13–2.99 (m, 2H), 2.76–2.52 (m, 3H), 2.46 (dq, J = 13.3, 5.9 Hz, 1H), 2.39−2.18 (m, 2H), 1.91–1.69 (m, 3H), 1.50–1.39 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.6, 146.8, 138.6, 130.6, 128.8, 127.9, 119.9, 70.1, 60.0, 59.1, 53.5, 52.9, 44.8, 40.3, 27.0, 26.9, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C18H22Cl2N3O2, 382.1084. Found 382.1070. HPLC purity: ≥99%, RT = 2.94.

(±)-(4aR,13bS)-10,11-dichloro-4-(2-isopropoxyethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one ((±)-trans-50b).
Steps 1–5:

See synthesis of (±)-trans-32b.

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(2-isopropoxyethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino-[5,6-b]quinazolin-8-one, (±)-Trans-50b.

Compound (±)-trans-50b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 2-(2-bromoethoxy)propane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 20 h. Purification by flash chromatography 3–20% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-50b as a tan solid (67.9 mg, 77%). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.74 (s, 1H), 4.18–4.08 (m, 2H), 3.65–3.46 (m, 3H), 3.07–3.00 (m, 1H), 2.96 (dt, J = 13.9, 5.8 Hz, 1H), 2.75 (dt, J = 13.5, 6.4 Hz, 1H), 2.70–2.61 (m, 2H), 2.55 (dq, J = 13.3, 5.5 Hz, 1H), 2.38 (dtd, J = 23.7, 12.0, 10.9, 3.9 Hz, 2H), 1.90–1.65 (m, 3H), 1.50–1.38 (m, 1H), 1.15 (dd, J = 6.1, 1.2 Hz, 6H). 13C NMR (101 MHz, CDCl3) δ 160.6, 157.6, 146.8, 138.6, 130.6, 128.8, 127.9, 119.9, 72.1, 65.3, 59.8, 53.5, 53.0, 45.2, 40.7, 27.3, 26.8, 25.1, 22.3, 22.2. HRMS (ESI) m/z: [M + H]+ Calcd for C20H26Cl2N3O2, 410.1397. Found 410.1397. HPLC purity: ≥99%, RT = 2.91 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(2-(dimethylamino)ethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-51b.
Steps 1–6:

See Synthesis of (±)-trans-48b

Step 7: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(2-(dimethylamino)ethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-trans-51b.

Compound (±)-trans-51b was prepared according to general procedure 9 by suspending (±)-trans-48b (1 equiv) and Et3N (2 equiv) in anhydrous DCE (0.25 M) and cooled to 0 °C. At 0 °C, MsCl (1.2 equiv) was added dropwise. Then allowed to warm to room temperature and stir for 2 h. After 2 h, dimethylamine (2 M in THF, 6 equiv) was added dropwise. The reaction was allowed to stir at room temperature for 22 h. Purification by flash chromatography 3–25% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-51b as an orange solid (33.2 mg, 44%).1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.74 (s, 1H), 4.24–4.04 (m, 2H), 3.10 (ddd, J = 12.1, 5.8, 3.4 Hz, 2H), 2.72–2.53 (m, 4H), 2.50–2.41 (m, 2H), 2.38 (s, 6H), 2.27 (td, J = 10.0, 5.1 Hz, 1H), 2.17 (td, J = 12.0, 2.9 Hz, 1H), 1.94–1.80 (m, 2H), 1.72 (qt, J = 13.0, 3.8 Hz, 1H), 1.50–1.37 (m, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.4, 146.8, 138.6, 130.6, 128.8, 128.8, 127.9, 119.8, 60.1, 56.6, 53.5, 51.0, 45.7, 44.9, 40.4, 27.1, 26.9, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C19H25Cl2N4O, 395.1400. Found 395.1415. HPLC Purity: ≥98%, RT = 2.99 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(2-(isopropylamino)ethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-52b.
Steps 1–6:

See Synthesis of (±)-Trans-48b

Step 7: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(2-(isopropylamino)ethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-52b.

Compound (±)-trans-52b was prepared according to procedure 11 by suspending (±)-trans-48b (1 equiv) and Et3N (2 equiv) in anhydrous DCE (0.25 M) and cooled to 0 °C. At 0 °C, MsCl (1.2 equiv) was added dropwise. Then allowed to warm to rt, stirred for 2 h, and then isopropylamine (6 equiv) was added dropwise. The reaction was allowed to stir at rt for 22 h. Purification by flash chromatography 75–100% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-52b as an orange solid (38.1 mg, 49%). 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.75 (s, 1H), 4.20 (ddd, J = 14.4, 8.4, 6.3 Hz, 1H), 4.05 (ddd, J = 14.9, 6.8, 5.4 Hz, 1H), 3.12–2.98 (m, 2H), 2.86–2.60 (m, 5H), 2.48–2.40 (m, 1H), 2.34–2.21 (m, 2H), 2.08 (td, J = 12.1, 2.7 Hz, 1H), 1.91–1.77 (m, 3H), 1.70 (ddt, J = 16.6, 12.9, 6.6 Hz, 1H), 1.52–1.40 (m, 1H), 1.09 (dd, J = 6.4, 3.4 Hz, 6H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.6, 146.8, 138.6, 130.6, 128.9, 127.9, 119.9, 60.3, 53.0, 52.9, 49.2, 44.9, 44.6, 40.3, 27.1, 24.9, 23.1, 22.9. HRMS (ESI) m/z: [M + H]+ Calcd for C20H27Cl2N4O, 409.1556. Found 409.1576. HPLC purity: ≥99%, RT = 3.02 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(oxetan-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-53b.
Steps 1–5:

See synthesis of (±)-trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(oxetan-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-53b.

Compound (±)-trans-53b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 3-(bromomethyl)-oxetane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 80 °C for 19 h. Purification by flash chromatography 3–30% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-53b as a tan solid (60.3 mg, 71%). 1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.73 (s, 1H), 4.81 (q, J = 7.7, 7.3 Hz, 2H), 4.44 (t, J = 5.9 Hz, 1H), 4.37 (t, J = 5.8 Hz, 1H), 4.21 (dt, J = 14.6, 7.1 Hz, 1H), 4.07 (dt, J = 14.0, 6.2 Hz, 1H), 3.24 (q, J = 8.2 Hz, 2H), 2.88 (d, J = 11.3 Hz, 1H), 2.64 (d, J = 12.3 Hz, 2H), 2.46 (dq, J = 19.2, 6.3, 5.8 Hz, 2H), 2.21 (s, 1H), 2.01 (t, J = 12.0 Hz, 1H), 1.85 (t, J = 14.3 Hz, 2H), 1.67 (d, J = 13.3 Hz, 1H), 1.49 (d, J = 3.8 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.3, 146.7, 138.7, 130.7, 128.9, 127.9, 119.8, 76.4, 76.4, 60.3, 56.8, 53.0, 45.9, 44.6, 40.1, 33.3, 26.9, 24.7. HRMS (ESI) m/z: [M + H]+ Calcd for C19H22Cl2N4O2, 394.1064. Found 394.1070. HPLC purity: ≥99%, RT = 2.85 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-((tetrahydro-2H-pyran-4-yl)-methyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-54b.
Steps 1–5:

See synthesis of (±)-Trans-32b

Step 6: Synthesis of (±)- (4aR,13bS)-10,11-Dichloro-4-((tetrahydro-2H-pyran-4-yl)methyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-54b.

Compound (±)-trans-54b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 4-bromomethyl-tetrahydropyran (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 80 °C for 20 h. Purification by flash chromatography 1–12% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-54b as a yellowish tan solid (109 mg, 70%).1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.75 (s, 1H), 4.20 (ddd, J = 14.4, 8.3, 6.2 Hz, 1H), 4.10–3.94 (m, 3H), 3.39 (tdd, J = 11.7, 9.2, 2.2 Hz, 2H), 3.10–3.04 (m, 1H), 2.67–2.57 (m, 3H), 2.38 (dq, J = 13.4, 5.9 Hz, 1H), 2.17 (td, J = 9.8, 5.2 Hz, 1H), 2.00 (ddd, J = 12.5, 10.5, 7.0 Hz, 2H), 1.84–1.56 (m, 6H), 1.50–1.39 (m, 1H), 1.32–1.18 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 160.5, 157.7, 146.8, 138.6, 130.6, 128.9, 127.9, 119.9, 68.1, 68.0, 60.6, 59.8, 53.9, 45.0, 40.3, 34.0, 32.0, 32.0, 27.3, 27.1, 24.9. HRMS (ESI) m/z: [M + H]+ Calcd for C21H26Cl2N3O, 422.1397. Found 422.1418. HPLC Purity: ≥98%, RT = 2.76 min.

(±)-(4aR,13bS)-10,11-Dichloro-4-(2-morpholinoethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-55b.
Steps 1–5:

See synthesis of (±)-trans-32b

Step 6: Synthesis of (±)-(4aR,13bS)-10,11-Dichloro-4-(2-morpholinoethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino-[5,6-b]quinazolin-8-one ((±)-Trans-55b).

Compound (±)-trans-55b was prepared according to general procedure 7 by suspending (±)-trans-32b (1 equiv), 4-(2-chloroethyl)morpholine hydrochloride (8 equiv), and Et3N (12 equiv) in DMF/MeOH (1.7:1 DMF:MeOH, 0.08 M) and heating to 80 °C for 24 h. Purification by flash chromatography 30–80% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-55b as a tan solid (55.8 mg, 59%). 1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.74 (s, 1H), 4.19 (ddd, J = 14.5, 8.4, 6.1 Hz, 1H), 4.07 (dt, J = 14.4, 6.0 Hz, 1H), 3.70 (t, J = 4.7 Hz, 4H), 3.10–3.04 (m, 1H), 3.02–2.91 (m, 1H), 2.65 (ddt, J = 12.0, 9.5, 3.6 Hz, 2H), 2.55–2.41 (m, 8H), 2.28 (td, J = 10.5, 5.2 Hz, 1H), 2.21 (td, J = 12.0, 2.9 Hz, 1H), 1.89–1.80 (m, 2H), 1.70 (qt, J = 13.1, 3.8 Hz, 1H), 1.45 (tdd, J = 14.0, 10.8, 6.6 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.4, 157.4, 146.8, 138.7, 130.6, 128.9, 127.9, 119.8, 67.0, 59.9, 56.3, 54.4, 53.6, 50.6, 45.0, 40.3, 27.1, 26.9, 24.9. HRMS (ESI) m/z: [M + H]+ Calcd for C21H27Cl2N4O2, 437.1506. Found 437.1509. HPLC Purity: ≥98%, RT = 2.85 min.

(±)-(4aR,13bS)-10-Chloro-4-(2-hydroxyethyl)-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-56b.
Steps 1–5:

See Synthesis of (±)-Trans-33b

Step 6: Synthesis of (±)-(4aR,13bS)-10-Chloro-4-(2-hydroxyethyl)-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-56b.

Compound (±)-trans-56b was prepared according to general procedure 7 by suspending (±)-trans-33b (1 equiv), 2-bromoethanol (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 24 h. Purification by flash chromatography 0.5–10% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-56b as a tan solid (484 mg, 73%). 1H NMR (400 MHz, CDCl3) δ 8.19 (s, 1H), 7.04 (s, 1H), 4.20 (ddd, J = 14.3, 8.2, 5.9 Hz, 1H), 4.05 (dt, J = 14.4, 5.9 Hz, 1H), 3.99 (s, 3H), 3.73 (ddd, J = 10.9, 9.2, 3.8 Hz, 1H), 3.59 (dt, J = 10.9, 4.4 Hz, 1H), 3.12 (ddt, J = 18.2, 9.2, 4.2 Hz, 2H), 2.71–2.60 (m, 3H), 2.46–2.33 (m, 2H), 2.29 (dt, J = 13.2, 3.7 Hz, 1H), 2.15 (td, J = 12.1, 2.8 Hz, 1H), 1.92–1.77 (m, 2H), 1.70 (qt, J = 13.0, 3.7 Hz, 1H), 1.51 (qd, J = 12.7, 3.5 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.5, 159.6, 156.8, 148.4, 127.7, 122.5, 114.3, 108.1, 60.4, 58.7, 56.7, 54.1, 53.0, 45.0, 39.9, 27.2, 27.2, 24.9. HRMS (ESI) m/z: [M + H]+ Calcd for C18H23ClN3O3, 364.1422. Found 364.1414. HPLC Purity: ≥97%, RT = 2.09 min.

(±)-(4aR,13bS)-10-Chloro-11-methoxy-4-(oxetan-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-57b.
Steps 1–5:

See synthesis of (±)-trans-33b

Step 6: Synthesis of (±)-(4aR,13bS)-10-Chloro-11-methoxy-4-(oxetan-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-57b.

Compound (±)-trans-57b was prepared according to general procedure 7 by suspending (±)-trans-33b (1 equiv), 3-(bromomethyl)-oxetane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 80 °C for 20 h. Purification by flash chromatography 20–40% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-57b as a white solid (48.1 mg, 57%). 1H NMR (400 MHz, CDCl3) δ 8.21 (s, 1H), 7.05 (s, 1H), 4.81 (ddd, J = 9.1, 7.4, 6.0 Hz, 2H), 4.44 (t, J = 6.0 Hz, 1H), 4.38 (t, J = 5.9 Hz, 1H), 4.20 (ddd, J = 14.5, 8.4, 6.1 Hz, 1H), 4.08 (dt, J = 14.4, 6.1 Hz, 1H), 3.99 (s, 3H), 3.31–3.19 (m, 2H), 2.88 (dt, J = 11.4, 3.8 Hz, 1H), 2.70–2.59 (m, 2H), 2.53–2.40 (m, 2H), 2.19 (td, J = 9.9, 5.2 Hz, 1H), 2.00 (td, J = 11.9, 2.8 Hz, 1H), 1.88–1.77 (m, 2H), 1.67 (qt, J = 13.0, 3.6 Hz, 1H), 1.47 (qd, J = 13.9, 13.5, 3.5 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 156.9, 148.4, 127.7, 122.5, 114.3, 108.1, 76.5, 76.5, 60.4, 56.8, 56.7, 53.1, 44.7, 40.0, 33.4, 27.2, 27.1, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C20H25ClN3O3, 390.1579. Found 390.1589. HPLC Purity: ≥99%, RT = 2.10 min.

(4aR,13bS)-10-Chloro-11-methoxy-4-(oxetan-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, Trans-57b-ent-1 and (4SR,13bR)-10-Chloro-11-methoxy-4-(oxetan-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-57b-ent-2.

500 mg of (±)-trans-57b was separated by Lotus Separations (Princeton University, Princeton, NJ) using chiral supercritical fluid chromatography. Analytical SFC Conditions: Agilent 1260 SFC equipped with a Diacel ChirralCel OJ-H, 250 (L) × 4.6 (ID) mm column. Temperature-Ambient. Mobile Phase CO2 Modifier: 30% EtOH with 0.1% diethylamine. Flow rate: 2 mL/min. Back Pressure: 100 bar. UV wavelength: 220 nm.

Preparative SFC Conditions.

Berger Multi-Gram-II Preparative-SFC equipped with a Diacel ChiralCel OJ-H, 250 (L) × 20 (ID) mm. Temperature: 35 °C. CO2 Modifier: 25% EtOH with 0.1% diethylamine. Flow rate: 45 mL/min. Back Pressure: 100 bar. UV wavelength: 220 nm.

(4aR,13bS)-10-Chloro-11-methoxy-4-(oxetan-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, Trans-57b-ent-1.

235 mg isolated from the chiral separation (>99% ee, RT = 4.68 min). Absolute stereochemistry determined by X-ray crystallography. 1H NMR (400 MHz, CDCl3) δ 8.21 (s, 1H), 7.05 (s, 1H), 4.81 (ddd, J = 9.1, 7.4, 6.0 Hz, 2H), 4.44 (t, J = 6.0 Hz, 1H), 4.38 (t, J = 5.9 Hz, 1H), 4.20 (ddd, J = 14.5, 8.4, 6.1 Hz, 1H), 4.08 (dt, J = 14.4, 6.1 Hz, 1H), 3.99 (s, 3H), 3.31–3.19 (m, 2H), 2.88 (dt, J = 11.4, 3.8 Hz, 1H), 2.70–2.59 (m, 2H), 2.53–2.40 (m, 2H), 2.19 (td, J = 9.9, 5.2 Hz, 1H), 2.00 (td, J = 11.9, 2.8 Hz, 1H), 1.88–1.77 (m, 2H), 1.67 (qt, J = 13.0, 3.6 Hz, 1H1.46 (qd, J = 14.0, 13.6, 3.7 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 156.9, 148.4, 127.7, 122.5, 114.3, 108.1, 76.5, 76.5, 60.4, 56.8, 56.7, 53.1, 44.7, 40.0, 33.4, 27.2, 27.1, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C20H25ClN3O3, 390.1579. Found 390.1589. HPLC Purity: ≥99%, RT = 2.10 min.

(4SR,13bR)-10-Chloro-11-methoxy-4-(oxetan-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, Trans-57b-ent-2.

225 mg isolated from the chiral separation (>99% ee, RT = 6.67 min). Absolute stereochemistry determined by X-ray crystallography. 1H NMR (400 MHz, CDCl3) δ 8.20 (s, 1H), 7.04 (s, 1H), 4.81 (ddd, J = 9.1, 7.4, 6.0 Hz, 2H), 4.44 (t, J = 6.0 Hz, 1H), 4.37 (t, J = 5.9 Hz, 1H), 4.20 (ddd, J = 14.5, 8.4, 6.1 Hz, 1H), 4.08 (dt, J = 14.4, 6.1 Hz, 1H), 3.99 (s, 3H), 3.30–3.19 (m, 2H), 2.87 (dt, J = 11.7, 3.6 Hz, 1H), 2.68–2.59 (m, 2H), 2.53–2.39 (m, 2H), 2.19 (td, J = 9.9, 5.2 Hz, 1H), 2.00 (td, J = 11.9, 2.8 Hz, 1H), 1.89–1.77 (m, 2H), 1.66 (qt, J = 13.1, 3.9 Hz, 1H), 1.46 (qd, J = 14.2, 13.7, 3.8 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.5, 159.6, 156.8, 148.4, 127.7, 122.5, 114.3, 108.0, 76.5, 76.5, 60.4, 56.8, 56.7, 53.0, 44.7, 40.0, 33.4, 27.2, 27.1, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C20H25ClN3O3, 390.1579. Found 390.1587. HPLC purity: ≥99%, RT = 2.10 min.

(±)-(4aR,13bS)-11-Methoxy-4-(oxetan-3-ylmethyl)-8-oxo-2,3,4,4a,5,6,8,13b-octahydro-1H-[1,6]naphthyridino[5,6-b]-quinazoline-10-carbonitrile ((±)-Trans-58b).
Steps 1–6:

See synthesis of (±)-Trans-57

Step 7: Synthesis of (±)-(4aR,13bS)-11-Methoxy-4-(oxetan-3-ylmethyl)-8-oxo-2,3,4,4a,5,6,8,13b-octahydro-1H-[1,6]-naphthyridino[5,6-b]quinazoline-10-carbonitrile, (±)-Trans-58b.

Compound (±)-trans-58b was prepared from (±)-trans-57b. (±)-trans-57b (1 equiv), Pd2(dba)3 (10 mol %), S-Phos (20 mol %), and Zn(CN)2 (1.2 equiv) were combined in a 2–5 mL MWI tube which was sealed, evacuated, and backfilled with Ar(g) two times. Then DMF/H2O (99:1, 0.11 M) was added and the tube was heated MWI 120 °C for 1.5 h. The reaction mixture was concentrated, and the residue was suspended MeOH (20 mL) and absorbed to a 10 g HSCX column (Agilent). After rinsing the column with an additional 80 mL of MeOH, the crude material was then collected by flushing the column with 60 mL 7N NH3/MeOH. Purification by flash chromatography 0.5–10% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-58b as a yellow-white solid (72.9 mg, 50%). 1H NMR (400 MHz, CDCl3) δ 8.43 (s, 1H), 7.03 (s, 1H), 4.81 (dt, J = 9.5, 6.5 Hz, 2H), 4.43 (t, J = 5.8 Hz, 1H), 4.37 (t, J = 5.8 Hz, 1H), 4.18 (ddd, J = 14.5, 8.6, 6.2 Hz, 1H), 4.13–4.05 (m, 1H), 4.01 (s, 3H), 3.29–3.18 (m, 2H), 2.88 (d, J = 11.3 Hz, 1H), 2.72–2.61 (m, 2H), 2.55–2.41 (m, 2H), 2.22 (q, J = 8.9 Hz, 1H), 2.01 (t, J = 11.8 Hz, 1H), 1.91–1.78 (m, 2H), 1.67 (q, J = 13.7 Hz, 1H), 1.46 (qd, J = 15.5, 14.4, 5.4 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 164.1, 160.1, 159.5, 152.4, 134.3, 115.6, 114.1, 107.5, 101.8, 76.4, 76.4, 60.2, 56.8, 56.7, 53.0, 44.9, 40.1, 33.4, 27.1, 26.9, 24.7. HRMS (ESI) m/z: [M + H]+ Calcd for C21H25ClN4O3, 381.1921. Found 381.1911. HPLC purity: ≥99%, RT = 2.49 min.

(±)-(4aR,13bS)-10-Chloro-4-(cyclobutylmethyl)-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-59b.
Steps 1–5:

See synthesis of (±)-trans-33b.

Step 6: Synthesis of (±)-(4aR,13bS)-10-Chloro-4-(cyclobutyl-methyl)-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-59b.

Compound (±)-trans-59b was prepared according to general procedure 7 by suspending (±)-trans-33b (1 equiv), (bromomethyl)cyclobutane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 80 °C for 20 h. Purification by flash chromatography 0.5–15% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-59b as a yellow-white solid (217 mg, 72%). 1H NMR (400 MHz, CDCl3) δ 8.19 (s, 1H), 7.04 (s, 1H), 4.18 (ddd, J = 14.5, 8.4, 6.1 Hz, 1H), 4.06 (dt, J = 14.3, 6.0 Hz, 1H), 3.98 (s, 3H), 3.00 (dt, J = 11.6, 3.6 Hz, 1H), 2.84 (dd, J = 13.1, 7.0 Hz, 1H), 2.70–2.50 (m, 3H), 2.47–2.29 (m, 2H), 2.19 (td, J = 9.9, 5.2 Hz, 1H), 2.06 (qdd, J = 12.8, 11.7, 6.8, 3.5 Hz, 3H), 1.98–1.60 (m, 7H), 1.45 (dd, J = 13.3, 3.4 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 157.2, 148.4, 127.7, 122.4, 114.3, 108.0, 60.0, 59.8, 56.6, 53.1, 44.8, 40.1, 33.6, 28.7, 28.0, 27.3, 26.9, 25.0, 18.9. HRMS (ESI) m/z: [M + H]+ Calcd for C21H27ClN3O2, 388.1786. Found 388.1781. HPLC purity: ≥99%, RT = 2.77 min.

(±)-(4aR,13bS)-10-Chloro-4-(cyclopropylmethyl)-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-60b.
Steps 1–5:

See Synthesis of (±)-Trans-33b

Step 6: Synthesis of (±)-(4aR,13bS)-10-Chloro-4-(cyclopropyl-methyl)-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-60b.

Compound (±)-trans-60b was prepared according to general procedure 7 by suspending (±)-trans-33b (1 equiv), (bromomethyl)-cyclopropane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 80 °C for 20 h. Purification by flash chromatography 2–20% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-60b as a yellow solid (358 mg, 70%).1H NMR (400 MHz, CDCl3) δ 8.20 (s, 1H), 7.05 (s, 1H), 4.19 (ddd, J = 14.5, 8.4, 6.1 Hz, 1H), 4.07 (dt, J = 14.4, 6.1 Hz, 1H), 3.99 (s, 3H), 3.29 (dt, J = 11.9, 2.8 Hz, 1H), 2.77 (dd, J = 13.4, 6.1 Hz, 1H), 2.69 (td, J = 12.3, 11.6, 3.7 Hz, 2H), 2.42 (dq, J = 13.3, 5.9 Hz, 1H), 2.33–2.20 (m, 3H), 1.95–1.67 (m, 3H), 1.48 (qd, J = 13.6, 13.1, 3.8 Hz, 1H), 0.95–0.83 (m, 1H), 0.60–0.47 (m, 2H), 0.16–0.08 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 157.2, 148.4, 127.7, 122.4, 114.3, 108.1, 59.6, 58.4, 56.7, 53.1, 44.9, 40.1, 27.3, 26.8, 25.1, 7.4, 4.9, 3.5. HRMS (ESI) m/z: [M + H]+ Calcd for C20H25ClN3O2, 374.1611. Found 374.1622. HPLC purity: ≥99%, RT = 2.43 min.

(±)-(4aR,13bS)-10-Chloro-4-(2-isopropoxyethyl)-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-61b.
Steps 1–5:

See synthesis of (±)-Trans-33b

Step 6: Synthesis of (±)-(4aR,13bS)-10-Chloro-4-(2-isopropoxyethyl)-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-61b.

Compound (±)-trans-61b was prepared according to general procedure 7 by suspending (±)-trans-33b (1 equiv), 1-bromo-2-methylpropane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 50 °C for 20 h. Purification by flash chromatography 0.5–10% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-61b as yellow solid (193 mg, 61%).1H NMR (400 MHz, CDCl3) δ 8.19 (s, 1H), 7.04 (s, 1H), 4.17–4.06 (m, 2H), 3.98 (s, 3H), 3.65–3.45 (m, 3H), 3.07–3.01 (m, 1H), 3.01–2.90 (m, 1H), 2.78–2.60 (m, 3H), 2.53 (dq, J = 13.2, 5.5 Hz, 1H), 2.37 (dtd, J = 19.6, 11.9, 10.9, 3.9 Hz, 2H), 1.89–1.65 (m, 3H), 1.45 (qd, J = 13.0, 4.2 Hz, 1H), 1.14 (dd, J = 6.1, 1.0 Hz, 6H). 13C NMR (101 MHz, CDCl3) δ 160.7, 159.6, 157.0, 148.4, 127.7, 122.4, 114.3, 108.0, 72.0, 65.3, 59.9, 56.6, 53.5, 53.0, 45.1, 40.5, 27.4, 26.8, 25.1, 22.3, 22.2. HRMS (ESI) m/z: [M + H]+ Calcd for C21H29ClN3O3, 406.1892. Found 406.1886. HPLC purity: ≥99%, RT = 3.25 min.

(±)-(4aR,13bS)-10-Chloro-4-isobutyl-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-62b.
Steps 1–5:

See Synthesis of (±)-Trans-33b

Step 6: Synthesis of (±)-(4aR,13bS)-10-Chloro-4-isobutyl-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6b]quinazolin-8-one, (±)-Trans-62b.

Compound (±)-trans-62b was prepared according to general procedure 7 by suspending (±)-trans-33b (1 equiv), 1-bromo-2-methylpropane (4 equiv), and Et3N (6 equiv) in DMF (0.25 M) and heating to 80 °C for 20 h. Purification by flash chromatography 0.5–10% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-62b as an off-white solid (205 mg, 70%). 1H NMR (400 MHz, CDCl3) δ 8.21 (s, 1H), 7.06 (s, 1H), 4.20 (ddd, J = 14.4, 8.3, 6.2 Hz, 1H), 4.05 (dt, J = 14.3, 6.1 Hz, 1H), 4.00 (s, 3H), 3.06 (d, J = 11.5 Hz, 1H), 2.70–2.60 (m, 2H), 2.47 (dd, J = 12.6, 9.2 Hz, 1H), 2.37 (dq, J = 13.4, 5.9 Hz, 1H), 2.15 (td, J = 9.9, 5.3 Hz, 1H), 2.02–1.87 (m, 2H), 1.87–1.61 (m, 4H), 1.47 (qd, J = 13.7, 13.2, 4.3 Hz, 1H), 0.91 (dd, J = 10.9, 6.5 Hz, 6H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 157.4, 148.5, 127.8, 122.4, 114.4, 108.1, 61.8, 60.9, 56.7, 53.7, 44.9, 40.1, 27.4, 26.9, 25.0, 21.4, 20.9. HRMS (ESI) m/z: [M + H]+ Calcd for C20H27ClN3O2, 376.1786. Found 376.1784. HPLC purity: ≥99%, RT = 3.05 min.

(4aR,13bS)-10-Chloro-4-isobutyl-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, trans-62b-ent-1 and (4aS,13bR)-10-chloro-4-isobutyl-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, Trans-62b-ent-2.

100 mg of (±)-trans-62b was separated under the following conditions: (R,R)-Welk-O 1 5 μm Kromasil, 25 cm × 21.1 mm: 5% isocratic gradient B/A (B = 2% IPA/DCM; A = 25% B in hexanes), 1.5 mL injection, 23 mL/min.

(4aR,13bS)-10-Chloro-4-isobutyl-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-62b-ent-1.

Absolute stereochemistry arbitrarily assigned. 42.1 mg isolated from the chiral separation (>99% ee, RT = 19.24 min). 1H NMR (400 MHz, CDCl3) δ 8.21 (s, 1H), 7.06 (s, 1H), 4.20 (ddd, J = 14.4, 8.3, 6.2 Hz, 1H), 4.05 (dt, J = 14.3, 6.1 Hz, 1H), 4.00 (s, 3H), 3.06 (d, J = 11.5 Hz, 1H), 2.70–2.60 (m, 2H), 2.47 (dd, J = 12.6, 9.2 Hz, 1H), 2.37 (dq, J = 13.4, 5.9 Hz, 1H), 2.15 (td, J = 9.9, 5.3 Hz, 1H), 2.02–1.87 (m, 2H), 1.87–1.61 (m, 4H), 1.47 (qd, J = 13.7, 13.2, 4.3 Hz, 1H), 0.91 (dd, J = 10.9, 6.5 Hz, 6H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 157.4, 148.5, 127.8, 122.4, 114.4, 108.1, 61.8, 60.9, 56.7, 53.7, 44.9, 40.1, 27.4, 26.9, 25.0, 21.4, 20.9. HRMS (ESI) m/z: [M + H]+ Calcd for C20H27ClN3O2, 376.1786. Found 376.1784. HPLC purity: ≥99%, RT = 3.05 min.

(4aS,13bR)-10-Chloro-4-isobutyl-11-methoxy-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]quinazolin-8-one, Trans-62b-ent-2.

Absolute stereochemistry arbitrarily assigned. 34.6 mg isolated from the chiral separation (>99% ee, RT = 20.86 min). 1H NMR (400 MHz, CDCl3) δ 8.21 (s, 1H), 7.06 (s, 1H), 4.20 (ddd, J = 14.4, 8.3, 6.1 Hz, 1H), 4.05 (dt, J = 14.3, 6.1 Hz, 1H), 4.00 (s, 3H), 3.07 (dd, J = 10.9, 4.1 Hz, 1H), 2.71–2.58 (m, 2H), 2.47 (dd, J = 12.6, 9.3 Hz, 1H), 2.37 (dq, J = 13.6, 5.9 Hz, 1H), 2.15 (td, J = 9.8, 5.2 Hz, 1H), 2.04–1.87 (m, 2H), 1.86–1.73 (m, 3H), 1.73–1.61 (m, 1H), 1.55–1.39 (m, 1H), 0.91 (dd, J = 11.0, 6.5 Hz, 6H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 157.4, 148.5, 127.8, 122.4, 114.4, 108.1, 61.8, 60.9, 56.7, 53.7, 44.9, 40.1, 27.4, 26.9, 25.0, 21.4, 20.9. HRMS (ESI) m/z: [M + H]+ Calcd for C20H27ClN3O2, 376.1786. Found 376.1782. HPLC purity: ≥99%, RT = 3.05 min.

(±)-(4aR,13bS)-10-Chloro-11-methoxy-4-(2-morpholinoethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-63b.
Steps 1–6:

See Synthesis of (±)-Trans-56b

Step 7: Synthesis of (±)-(4aR,13bS)-10-Chloro-11-methoxy-4-(2-morpholinoethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-63b.

Compound (±)-trans-63b was prepared according to procedure 11 by suspending (±)-trans-56b (1 equiv) and Et3N (2 equiv) in anhydrous DCE (0.25 M) and cooled to 0 °C. At 0 °C, MsCl (1.2 equiv) was added dropwise. The reaction was allowed to warm to rt and stirred for 2 h. Morpholine (8 equiv) was added dropwise and the mixture was stirred at rt for 23h and then heated to reflux for 7 h. Purification by flash chromatography 50–75% X/hexanes (X = 2% NEt3 3:1 EtOAc/EtOH) afforded (±)-trans-63b as a tan solid (143 mg, 40%). 1H NMR (400 MHz, CDCl3) δ 8.19 (s, 1H), 7.03 (s, 1H), 4.17 (ddd, J = 14.4, 8.4, 6.0 Hz, 1H), 4.07 (dt, J = 14.3, 6.0 Hz, 1H), 3.98 (s, 3H), 3.69 (q, J = 3.9, 3.2 Hz, 4H), 3.08 (dt, J = 11.7, 3.4 Hz, 1H), 3.02–2.92 (m, 1H), 2.70–2.60 (m, 2H), 2.55–2.39 (m, 8H), 2.25 (dtd, J = 27.4, 11.9, 10.8, 4.0 Hz, 2H), 1.89−1.79 (m, 2H), 1.71 (qt, J = 13.1, 3.7 Hz, 1H), 1.46 (qd, J = 13.4, 3.9 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 156.9, 148.4, 127.7, 122.5, 114.3, 108.0, 67.0, 60.1, 56.7, 56.3, 56.2, 54.3, 54.2, 53.6, 50.6, 44.9, 40.1, 27.3, 27.0, 25.0. HRMS (ESI) m/z: [M + H]+ Calcd for C22H30ClN4O3, 433.2001. Found 433.1998. HPLC purity: ≥98%, RT = 2.26 min.

(±)-(4aR,13bS)-10-Chloro-11-methoxy-4-(2-(pyrrolidin-1-yl)-ethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-64b.
Steps 1–6:

See synthesis of (±)-Trans-56b

Step 7: Synthesis of (±)-(4aR,13bS)-10-Chloro-11-methoxy-4-(2-(pyrrolidin-1-yl)ethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-64b.

Compound (±)-trans-64b was prepared according to general procedure 9 by suspending (±)-trans-56b (1 equiv) and Et3N (2 equiv) in anhydrous DCE (0.25 M). After cooling to 0 °C, MsCl (1.2 equiv) was added dropwise and then the reaction was allowed to warm to rt and stirred for 2 h. Pyrrolidine (8 equiv) was added dropwise. The reaction was allowed to stir at rt for 23 h and then heated to reflux for 7 h. Purification by flash chromatography 50–80% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-64b as a yellow-orange solid (24.7 mg, 17%). 1H NMR (400 MHz, CDCl3) δ 8.19 (s, 1H), 7.03 (s, 1H), 4.22–4.02 (m, 2H), 3.98 (s, 3H), 3.12–2.97 (m, 2H), 2.73–2.40 (m, 10H), 2.32–2.17 (m, 2H), 1.91–1.63 (m, 7H), 1.46 (qd, J = 13.2, 4.1 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.6, 159.6, 157.0, 148.4, 127.7, 122.4, 114.3, 108.0, 60.1, 60.0, 56.6, 54.7, 53.7, 53.6, 52.3, 44.9, 40.2, 27.3, 27.2, 27.0, 25.0, 23.5. HRMS (ESI) m/z: [M + H]+ Calcd for C22H39ClN4O2, 417.2052. Found 417.2053. HPLC purity: ≥99%, RT = 2.36 min.

(±)-(4aR,13bS)-10-Chloro-11-methoxy-4-(pyridin-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]naphthyridino[5,6-b]-quinazolin-8-one, (±)-Trans-65b.
Steps 1–5:

See Synthesis of (±)-Trans-56b

Step 6: Synthesis of (±)-(4aR,13bS)-10-Chloro-11-methoxy-4-(pyridin-3-ylmethyl)-1,2,3,4,4a,5,6,13b-octahydro-8H-[1,6]-naphthyridino[5,6-b]quinazolin-8-one, (±)-Trans-65b.

Compound (±)-trans-65b was prepared according to general procedure 7 by suspending (±)-trans-33b (1 equiv), 3-(bromomethyl)pyridine hydrobromide (1.2 equiv), and Et3N (3 equiv) in DMF (0.25 M) and heating to 50 °C for 24 h. Purification by flash chromatography 20–40% X/hexanes (X = 2% NEt3, 3:1 EtOAc/EtOH) afforded (±)-trans-65b as an off-white solid (143 mg, 37%). 1H NMR (400 MHz, CDCl3) δ 8.58 (d, J = 2.2 Hz, 1H), 8.53 (dd, J = 4.8, 1.6 Hz, 1H), 8.23 (s, 1H), 7.69 (dt, J = 7.9, 2.0 Hz, 1H), 7.32–7.24 (m, 1H), 7.07 (s, 1H), 4.30 (ddd, J = 14.3, 8.1, 6.3 Hz, 1H), 4.20–4.04 (m, 2H), 4.01 (s, 3H), 3.22 (d, J = 13.9 Hz, 1H), 2.90 (dt, J = 11.4, 3.7 Hz, 1H), 2.76 (td, J = 11.1, 3.7 Hz, 1H), 2.72–2.63 (m, 1H), 2.52 (dq, J = 12.3, 6.0 Hz, 1H), 2.33 (ddd, J = 10.7, 9.1, 5.4 Hz, 1H), 2.08–1.87 (m, 2H), 1.86–1.75 (m, 1H), 1.67 (qt, J = 12.8, 3.7 Hz, 1H), 1.52 (qd, J = 13.1, 3.9 Hz, 1H). 13C NMR (101 MHz, CDCl3) δ 160.5, 159.6, 156.9, 150.3, 148.8, 148.4, 136.4, 134.3, 127.7, 123.5, 122.5, 114.3, 108.1, 60.5, 56.7, 55.3, 53.2, 44.9, 39.8, 27.4, 27.2, 24.8. HRMS (ESI) m/z: [M + H]+ Calcd for C22H24ClN4O2, 411.1561. Found 411.1567. HPLC purity: ≥99%, RT = 3.21 min.

X-ray Crystallography.

CCDC 2370879 and CCDC 2370880 contain the supporting crystallographic data for compounds (4aR,13bS)-57b-ent-1 (BDGR-20236) and (4aS,13bR)-57b-ent-2 (BDGR-20237), respectively. These data can be obtained free of charge from The Cambridge Crystallographic Data Centre via www.ccdc.cam.ac.uk/structures. Data collection and refinement statistics are reported in the Supporting Information.

In Vitro Assay Experimental Protocols.
Amoeba Strains.

N. fowleri strain Nf69 (ATCC 30215) trophozoites, which were originally isolated from a patient in Australia in 1969, were chosen to perform dose response evaluations. N. fowleri strain TY (ATCC 30107) trophozoites, isolated from a patient in Virginia, USA in 1969, were used to score compounds for potential geographic differences in strain susceptibility. Both strains were generously donated by Dr. Dennis Kyle at UGA.

Determination of EC50 Values in the N. fowleri Assay.

N. fowleri strain Nf69 (ATCC 30215) trophozoites were cultured axenically as previously described.29 Briefly, amoeba were seeded at 1 × 104 cells/mL in 100 μL of the indicated medium in white TC-treated 96-well plates (Thermo Scientific Nunc Microwell 136101). Cells were seeded into Nelson’s Complete Media (NCM, 0.17% liver infusion broth (BD Difco, Franklin Lakes, NJ), 0.17% glucose, 0.012% sodium chloride, 0.0136% potassium phosphate monobasic, 0.0142% sodium phosphate dibasic, 0.0004% calcium chloride, 0.0002% magnesium sulfate, 10% heat-inactivated fetal bovine serum (FBS), 1% penicillin-streptomycin) at 37 °C in 96-well tissue culture plates. Vehicle or compound at concentrations initially ranging from 20 to 0.375 μM were added to cells, followed by culture for 48 h (37 °C, 5% CO2). Plates were equilibrated to rt for 15 min and CellTiter Glo reagent was added followed by orbital shaking for 2 min. After an additional incubation (rt, 10 min), luminescence was scored on a BioTek Synergy H1 microplate reader. Luminescence values were used to calculate percent cell growth inhibition (based on controls) for each concentration of compound tested. Averages of triplicates were calculated and fit to dose–response curves for the determination of EC50 values, using Prism 9.0 (GraphPad Software, San Diego, CA). Miltefosine (Sigma-Aldrich) was used a positive control (N. fowleri EC50 = 35.7 ± 9.10 μM). Other controls included media alone, cells and media, and vehicle and media. For very potent compounds, concentration of reagent in the dose–response assays were reduced to provide an appropriate range for EC50 value resolution.

Determination of CC50 Values in the SH-SY5Y Cytotoxicity Assay.

Dry powder compound was dissolved in DMSO to a final concentration of 20 mM. Serial dilutions were performed to provide a starting concentration of 20 μM in the assay. SH-SY5Y cells (human neuroblastoma cell line, ATCC CRL-2266) were plated at 2000 cells per well into a Corning 3765 plate using a ThermoFisher multidrop reagent dispenser in 50 μL of media and allowed to attach overnight at 37 °C. Using an Echo acoustic dispenser (Labcyte), 50 nL of the diluted compound (20, 1 μM or a series dilution to afford a dose response curve) was added in triplicate to the target cell plates. Doxorubicin at the same concentrations as the test compound was used as a positive control. After incubating for 72 h, cell viability was evaluated by adding 25 μL of Cell Titer-Glo reagent (Promega) per well, followed by incubation (10 min), and detection of the luminescent signal using a Pherostar plate reader (BMG). Percent viability was determined by comparing the percent signal reduction as compared to DMSO negative control. Data was analyzed by Collaborative Drug Discovery (CDD) data management software to generate data points and dose response curves.

ADME Experimental Protocols.
Log D Determination.

Compound (2 μL of a 10 mM DMSO stock solution) was transferred into tubes in duplicate, and to each tube was added phosphate buffer (pH 7.4)-1-octanol solution (pH 7.4). After shaking of the tubes for 1 h at rt, contents were centrifuged, and buffer layer samples and 1-octanol layer samples were aliquoted. The samples were diluted with equal volumes of labetalol and tolbutamide in 50% methanol) and analyzed by LC-MS/MS.

Kinetic Solubility Determination.

To the lower chamber of a mini-uniprep vial was added separate dilutions of compound prepared from DMSO stock solutions, followed by the appropriate volume of phosphate buffer (pH 7.4). After shaking for 24 h at rt, the sample was centrifuged for 30 min, and mini-unipreps were compressed to collect filtrates which were analyzed by HPLC.

Liver Microsomal Stability.

To incubation plates containing mouse microsomes (CD-1 mouse, Corning, cat. no. 452413) in phosphate buffer was added acetonitrile-diluted DMSO stock solution of compound with mixing. Samples were with and without NADPH cofactor. Plates were incubated at 37 °C for 60 min while shaking. At incremental time points ranging from 5 to 60 min, samples were quenched using equal concentrations of tolbutamide and labetalol. Samples were centrifuged, diluted with water, and supernatant was sampled for LC-MS/MS analysis. Controls included: testosterone, diclofenac, and propafenone.

Mouse Plasma Stability.

DMSO stock solutions of test compound and controls (enalapril maleate, bisacodyl, and procaine HCl) were prepared. To a 96-well plate containing plasma (CD-1 mouse, EDTA-K2 anticoagulant, minimum of 20 male donors) was added compound or control, and the mixtures were incubated at 37 °C for 0, 10, 30, 60, or 120 min. At the indicated incubation time, tolbutamide and labetalol in acetonitrile were added to precipitate protein. After shaking for 20 min and centrifuging for 20 min, supernatant was added to water, shaken for 10 min, and then samples were analyzed by LCMS using an internal standard.

Plasma Protein Binding.

A 96-well equilibrium dialysis device (HTDialysis LLC, cat. No. 1101) was employed using CD-1 male mouse plasma with EDTA-K2 anticoagulant. Warfarin was used as a positive control. An aliquot of the compound in phosphate buffer was transferred to the donor side of each dialysis well in triplicate, and the dialysis buffer was loaded to the receiver side of the well. The dialysis plate was incubated at 37 °C with 5% CO2 on a slow shaking platform for 4 h. Samples from each side of the dialysis device were transferred to new 96-well plates and diluted with a solution of tolbutamide, labetalol, and metformin in acetonitrile which contained an internal standard. After mixing and centrifuging samples for 20 min, supernatants were analyzed by LCMS/MS.

BBB Parallel Artificial Membrane Permeation Assay.27

A DMSO stock solution of test compound was diluted in PBS (pH 7.4) and then added to a 96-well donor plate. The positive controls, verapamil, clonidine, hydrocortisone and theophylline were purchased from Sigma-Aldrich. The filter membrane was coated with dodecane-diluted porcine polar brain lipid (PBL, Avanti Polar Lipids, cat no. 141101P), The filter plate was filled with PBS buffer, and then the filter plate was carefully put on the donor plate to form a “sandwich” with test compound solution on the bottom, artificial lipid membrane in the middle, and the acceptor PBS on the top. After incubating at 18 h at rt, the donor plate was removed, and the solution from donor and filter wells were transferred into the vials from which the concentration ratios were determined by HPLC in triplicate.

In Vivo Assay Experimental Protocols.
Ethics Statement.

All procedures were carried out in accordance with the PHS Policy on the Care and Use of Laboratory Animals and in accord with the CU PHS Assurance Number D16–00435 (A3737–01) under the approval of the Clemson University Institutional Animal Care and Use Committee (IACUC). All animal research programs and facilities have full accreditation from the Association for Assessment and Accreditation of Laboratory Animal Care, International (AAALAC). Euthanasia by heavy anesthetization followed by bilateral pneumothorax was used based on recommendations from the AVAM Guidelines for the Euthanasia of Animals.

Mouse Pharmacokinetics Protocol.

For single bolus 12 mg/kg dosing, six female CD-1 mice (Charles River, 24.6–28.4 g) were split into two groups of three mice each. The group receiving an oral dose of compound was fasted. Compound was dissolved in 10% NMP/20% cremophor EL/70% water to give a clear solution. Mice administered compound by IV received 11.1 mg/kg while mice administered compound by PO received 10.7 mg/kg. Plasma concentrations were analyzed at 0.5, 2, 4, 8, and 12 h in each group. Brains were collected from mice in each group at 12 h, tissue was homogenized with MeOH/PBS, and the supernatant of processed samples were analyzed by LCMS/MS (ACQUITY UPLC BEH C18 1.7 μm 2.1 mm × 50 mm column), flow rate: 0.55 mL/min, mobile phase A:0.1% NH3·H2O and 2 mM NH4OAc in water/ACN (v/v, 95:5) and mobile phase B: 0.1% NH3·H2O and 2 mM NH4OAc in ACN/water (v/v, 95:5). Plasma LLOQ: 1 ng/mL; brain homogenate LLOQ: 10 ng/g. Verapamil was used as an internal standard. For single bolus 50 mg/kg dosing, three, fasted, female CD-1 mice (Charles River, 27 g) were administered compound by IP injection (43.8 mg compound as 5 mg/mL in 10% DMSO/30% cremophor EL/60% water, homogeneous opaque suspension). Plasma samples were obtained at 2 and 4 h. Brain tissue was analyzed at 4 h. Identical conditions for LCMS/MS analysis of samples were used as described above. Data from each cohort (n = 3) was averaged. Clinical observations were normal during both studies.

Mouse Efficacy Protocol.

Eighteen CD-1 female mice (aged 35–40 days, Charles River) were infected by nasal instillation with 5 × 104 N. fowleri Nf69 strain trophozoites that had been isolated after passage through mice to ensure virulence. Mice were provided unlimited food and water throughout the duration of the experiment. The three groups consisted of equal numbers of rodents. Groups received either vehicle (10% 1-methyl-2-pyrrolidinone (NMP), 20% Cremophor) or test agent delivered by IP injection for 5 days starting 48 h after infection. BDGR-20237 (11 mg/kg) was administered BID (12 h apart), while amphotericin B (5 mg/kg) was injected QD. Rodents were monitored carefully and euthanized when symptoms of infection, including failure to respond to stimuli or seizures, were evident. After 14 days, surviving animals were sacrificed and brains were removed and analyzed to assess cure. Mice were considered cured if amoebae from isolated brains failed to grow in culture after 2 weeks. Median survival of vehicle treated animals was 5.4 days, compared to 6.1 days for BDGR-20237 treated mice. The p value for the analysis was 0.09.

Supplementary Material

SI

ACKNOWLEDGMENTS

The authors acknowledge funding from the National Institute of Allergy and Infectious Diseases (J.E.G., R21AI161232 for chemistry method development; J.C.M., R21AI175463 and the US National Institutes of Health Center for Biomedical Excellence (COBRE) grant under award number P20GM146584.) The authors would like to thank Dr. Dennis Kyle at UGA for providing the N. fowleri strains used in this study. X-ray diffraction was possible through funding from NSF Award CHE-1919350 in the UW-Madison Department of Chemistry. Synthesis efforts used instrumentation at the UW-Madison Analytical Instrumentation Center and the Medicinal Chemistry Center, both within the UW-Madison School of Pharmacy. We also thank the University of Wisconsin Carbone Cancer Center (UWCCC) Small Molecule Screening Facility, especially Song Guo and Spencer Erickson, for SH-SY5Y toxicity screening (support grant P30 CA014520).

ABBREVIATIONS USED

ADME

absorption-distribution-metabolism-excretion

BBB

blood–brain barrier

BID

twice daily dosing

B/P

brain-to-plasma ratio

CNS

central nervous system

HLM

human liver microsomes

IACUC

Institutional Animal Care and Use Committee

IN

intranasal

IP

intraperitoneal

IV

intravenous

LLOQ

lower limit of quantification

MLM

mouse liver microsomes

MTD

maximum tolerated dose

N. fowleri

Naegleria fowleri

PAM

primary amoebic meningoencephalitis

PAMPA

parallel artificial membrane permeability assay

PO

oral administration

QD

once daily dosing

SAR

structure–activity relationship

SFC

supercritical fluid chromatography

SI

selectivity index

SPR

structure–property relationship

Tx

treatment

Footnotes

The authors declare the following competing financial interest(s): J.E.G., M.S.L., and J.C.M. are co-inventors of a patent supported through the Wisconsin Alumni Research Foundation (WARF) on the quinazolinones disclosed herein.

ASSOCIATED CONTENT

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.4c01630.

Molecular strings (CSV)

Crystallographic data collection and refinement statistics, chiral SFC information, and HPLC traces for BDGR-20236 and BDGR-20237. 1H, 13C, and 19F NMR spectra of isolated intermediates and final compounds (PDF)

Complete contact information is available at: https://pubs.acs.org/10.1021/acs.jmedchem.4c01630

Contributor Information

Matthew S. Lish, Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States

Jillian E. M. McKeon, Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina 29634, United States

Caroline M. Palmentiero, Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina 29634, United States

Julia M. Pomeroy, Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States

Colm P. Roster, Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina 29634, United States

Ilia A. Guzei, Molecular Structure Laboratory, Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States

James C. Morris, Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina 29634, United States

Jennifer E. Golden, Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States; School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States

REFERENCES

  • (1).Zamzuri MAIA; Majid FNA; Mihat M; Ibrahim SS; Ismail M; Aziz SA; Mohamed Z; Rejali L; Yahaya H; Abdullah Z; et al. Systematic Review of Brain-Eating Amoeba: A Decade Update. Int. J. Environ. Res. Public Health 2023, 20 (4), No. 3021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (2).Prevention, U. S.; Centers for Disease Control and Prevention. Naegleria fowleri Infection 2024. https://www.cdc.gov/naegleria/about/index.html. (accessed July 6, 2024). [Google Scholar]
  • (3).Yoder JS; Straif-Bourgeois S; Roy SL; Moore TA; Visvesvara GS; Ratard RC; Hill VR; Wilson JD; Linscott AJ; Crager R; et al. Primary amebic meningoencephalitis deaths associated with sinus irrigation using contaminated tap water. Clin. Infect. Dis. 2012, 55 (9), e79–e85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (4).Shaukat A; Khaliq N; Riaz R; Munsab R; Ashraf T; Raufi N; Shah H Noninvasive diagnostic biomarkers, genomic profiling, and advanced microscopic imaging in the early detection and characterization of Naegleria fowleri infections leading to primary amebic meningoencephalitis (PAM). Ann. Med. Surg. 2024, 86 (4), 2032–2048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (5).Baig AM Primary Amoebic Meningoencephalitis: Neurochemotaxis and Neurotropic Preferences of Naegleria fowleri. ACS Chem. Neurosci. 2016, 7 (8), 1026–1029. [DOI] [PubMed] [Google Scholar]
  • (6).Grace E; Asbill S; Virga K Naegleria fowleri: pathogenesis, diagnosis, and treatment options. Antimicrob. Agents Chemother. 2015, 59 (11), 6677–6681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (7).Marciano-Cabral F; Cabral GA The immune response to Naegleria fowleri amebae and pathogenesis of infection. FEMS Immunol. Med. Microbiol. 2007, 51 (2), 243–259. [DOI] [PubMed] [Google Scholar]
  • (8).Jarolim KL; McCosh JK; Howard MJ; John DT A light microscopy study of the migration of Naegleria fowleri from the nasal submucosa to the central nervous system during the early stage of primary amebic meningoencephalitis in mice. J. Parasitol. 2000, 86 (1), 50–55. [DOI] [PubMed] [Google Scholar]
  • (9).Güémez A; García E Primary Amoebic Meningoencephalitis by Naegleria fowleri: Pathogenesis and Treatments. Biomolecules 2021, 11 (9), No. 1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (10).Vugia DJR; Tarro J; Vareechon T; Pannaraj C; Traub PS; Cope E; Balter JRS Notes from the Field: Fatal Naegleria fowleri Meningoencephalitis After Swimming in Hot Spring Water California, 2018. Morb. Mortal. Wkly. Rep. 2019, 68 (36), 793–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (11).Maloney P; Mowrer C; Jansen L; Karre T; Bedrnicek J; Obaro SK; Iwen PC; McCutchen E; Wetzel C; Frederick J; et al. Fatal Primary Amebic Meningoencephalitis in Nebraska: Case Report and Environmental Investigation, August 2022. Am. J. Trop. Med. Hyg. 2023, 109 (2), 322–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (12).Maciver SK; Piñero JE; Lorenzo-Morales J Is Naegleria fowleri an Emerging Parasite? Trends Parasitol. 2020, 36 (1), 19–28. [DOI] [PubMed] [Google Scholar]
  • (13).Cope JR; Ali IK Primary Amebic Meningoencephalitis: What Have We Learned in the Last 5 Years? Curr. Infect. Dis. Rep. 2016, 18 (10), No. 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (14).Burqi AMK; Mahboob S; Anwar S; Bizanjo M; Rafique M; Rafique M; Ghanchi N Successful Treatment of Confirmed Naegleria fowleri Primary Amebic Meningoencephalitis. Emerging Infect. Dis. 2024, 30 (4), 803–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (15).Pugh JJ; Levy RA Naegleria fowleri: Diagnosis, Pathophysiology of Brain Inflammation, and Antimicrobial Treatments. ACS Chem. Neurosci. 2016, 7 (9), 1178–1179. [DOI] [PubMed] [Google Scholar]
  • (16).Baig AM; Khan NA Novel Chemotherapeutic Strategies in the Management of Primary Amoebic Meningoencephalitis Due to Naegleria fowleri. CNS Neurosci. Ther. 2014, 20 (3), 289–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (17).Cope JR; Conrad DA; Cohen N; Cotilla M; DaSilva A; Jackson J; Visvesvara GS Use of the Novel Therapeutic Agent Miltefosine for the Treatment of Primary Amebic Meningoencephalitis: Report of 1 Fatal and 1 Surviving Case. Clin. Infect. Dis. 2016, 62 (6), 774–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (18).Pomeroy JM; Khalifa MM; Milanes JE; Palmentiero CM; Morris JC; Golden JE Synthesis and Evaluation of Benzylamine Inhibitors of Neuropathogenic Naegleria fowleri “Brain-Eating” Amoeba. ACS Med. Chem. Lett. 2024, 15 (1), 87–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (19).Lish MS; Milanes JE; Sanders KM; Guzei IA; Morris JC; Golden JE Mannich-Type Condensation and Domino Quinazolinone-Amidine Rearrangement Affords Ring-Fused Mackinazolinones with Anti-Amoebic Activity. Adv. Synth. Catal. 2023, 365 (24), 4567–4575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (20).Fitz-Henley JN; Rozema SD; Golden JE Dihydropyrazinoquinazolinones via SN2 Sulfamidate Ring-Opening and a Sequential Quinazolinone–Amidine Rearrangement Strategy (SQuAReS). J. Org. Chem. 2022, 87 (21), 14889–14898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (21).Chobanian HR; Fors BP; Lin LS A facile microwave-assisted palladium-catalyzed cyanation of aryl chlorides. Tetrahedron Lett. 2006, 47 (19), 3303–3305. [Google Scholar]
  • (22).Olofson RA; Abbott DE Tests of a piperidino mask for the protection of functionalized carbon sites in multistep syntheses. J. Org. Chem. 1984, 49 (15), 2795–2799. [Google Scholar]
  • (23).Thavaneswaran S; McCamley K; Scammells PJ N-Demethylation of Alkaloids. Nat. Prod. Commun. 2006, 1 (10), No. 1934578X0600101008. [Google Scholar]
  • (24).Firth JD; O’Brien P; Ferris L Synthesis of Enantiopure Piperazines via Asymmetric Lithiation–Trapping of N-Boc Piperazines: Unexpected Role of the Electrophile and Distal N-Substituent. J. Am. Chem. Soc. 2016, 138 (2), 651–659. [DOI] [PubMed] [Google Scholar]
  • (25).Zyserman I; Mondal D; Sarabia F; McKerrow JH; Roush WR; Debnath A Identification of cysteine protease inhibitors as new drug leads against Naegleria fowleri. Exp. Parasitol. 2018, 188, 36–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (26).Jones LH; Summerhill NW; Swain NA; Mills JE Aromatic chloride to nitrile transformation: medicinal and synthetic chemistry. MedChemComm 2010, 1 (5), 309–318. 10.1039/C0MD00135J [DOI] [Google Scholar]
  • (27).Di L; Kerns EH; Bezar IF; Petusky SL; Huang Y Comparison of blood-brain barrier permeability assays: in situ brain perfusion, MDR1-MDCKII and PAMPA-BBB. J. Pharm. Sci. 2009, 98 (6), 1980–1991. [DOI] [PubMed] [Google Scholar]
  • (28).Humphrey JYE; Fisher E; Pecora A; Tarantino K Facile N-Cbz cleavage by stoichiometric sulfonic acid in HFIP DOI: 10.1021/scimeetings.2c00065. [DOI] [Google Scholar]
  • (29).Milanes JE; Suryadi J; Abendroth J; Van Voorhis WC; Barrett KF; Dranow DM; Phan IQ; Patrick SL; Rozema SD; Khalifa MM; et al. Enzymatic and Structural Characterization of the Naegleria fowleri Glucokinase. Antimicrob. Agents Chemother. 2019, 63 (5), No. 10–1128, DOI: 10.1128/AAC.02410-18. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

SI

RESOURCES